1
|
Sandforth L, Kullmann S, Sandforth A, Fritsche A, Jumpertz-von Schwartzenberg R, Stefan N, Birkenfeld AL. Prediabetes remission to reduce the global burden of type 2 diabetes. Trends Endocrinol Metab 2025:S1043-2760(25)00004-9. [PMID: 39955249 DOI: 10.1016/j.tem.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/12/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Prediabetes is a highly prevalent and increasingly common condition affecting a significant proportion of the global population. The heterogeneous nature of prediabetes presents a challenge in identifying individuals who particularly benefit from lifestyle or other therapeutic interventions aiming at preventing type 2 diabetes (T2D) and associated comorbidities. The phenotypic characteristics of individuals at risk for diabetes are associated with both specific risk profiles for progression and a differential potential to facilitate prediabetes remission and reduce the risk of future T2D. This review examines the current definition and global prevalence of prediabetes and evaluates the potential of prediabetes remission to reduce the alarming increase in the global burden of T2D.
Collapse
Affiliation(s)
- Leontine Sandforth
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Arvid Sandforth
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Reiner Jumpertz-von Schwartzenberg
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany; M3 Research Center, Malignom, Metabolome, Microbiome, 72076 Tübingen, Germany; Cluster of Excellence EXC 2124 'Controlling Microbes to Fight Infections' (CMFI), University of Tübingen, Tübingen, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany; Department of Diabetes, Life Sciences, and Medicine, Cardiovascular Medicine and Life Sciences, King's College London, London, UK.
| |
Collapse
|
2
|
Cinti F, Mezza T, Severi I, Moffa S, Giuseppe GD, Capece U, Ciccarelli G, Soldovieri L, Brunetti M, Morciano C, Gugliandolo S, Senzacqua M, Avolio A, Quero G, Tondolo V, Nista EC, Moroni R, Cinti S, Alfieri S, Gasbarrini A, Pontecorvi A, Giaccari A. In humans increase in intrapancreatic adipose tissue predicts beta-cell dedifferentiation score before diabetes onset: A pilot study. Diabetes Res Clin Pract 2025; 221:112029. [PMID: 39938572 DOI: 10.1016/j.diabres.2025.112029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/18/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND The role of intrapancreatic fat (WAT) in the development of T2D remains debated. In T2D, β-cell dedifferentiation is one of the mechanisms responsible for β-cell failure but its role in prediabetes is unknown. We aimed to investigate the relation between WAT and β-cell dedifferentiation prior to diabetes onset. METHODS We evaluated pancreatic samples from patients without history of diabetes, who had previously undergone an oral glucose tolerance test and hyperglycemic clamp. Subjects were divided into 3 glucose tolerance groups: normal (NGT), altered (IGT) or newly diagnosed diabetes (nDM). Dedifferentiation and WAT% were morphologically assessed. RESULTS WAT was higher in nDM patients compared to NGT and IGT (WAT nDM 43.79 ± 20.83 %, IGT 10.67 ± 8.5 %, NGT 4.43 ± 4.37 %). We observed a progressive increase in dedifferentiation score, in parallel with worsening glucose tolerance (from NGT to IGT to nDM; 4.8 ± 3.8; 32.37 ± 7.4; 40.38 ± 19 respectively). A strong linear regression established that WAT could statistically significantly predict dedifferentiated β-cells (R = 0.86, p = 0.005), and that the predicted increase in dedifferentiated β-cells was 1.25 points for every extra one-point change in WAT. Interestingly, the WAT and dedifferentiation score variable pair were significantly related to 1-hour post-load glycemia. CONCLUSIONS The accumulation of WAT might be responsible for dedifferentiation, making it a potential new target to curb diabetes onset.
Collapse
Affiliation(s)
- Francesca Cinti
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Teresa Mezza
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy; Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | - Ilenia Severi
- Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | - Simona Moffa
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Gianfranco Di Giuseppe
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Umberto Capece
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Gea Ciccarelli
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Laura Soldovieri
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Michela Brunetti
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Cassandra Morciano
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy; Dipartimento di Scienze Cliniche e Sperimentali, Medicina Interna - Università degli Studi di Brescia Brescia BS Italy
| | - Shawn Gugliandolo
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Martina Senzacqua
- Department of Clinical and Experimental Medicine Center of Obesity Università Politecnica delle Marche Ancona Italy
| | - Adriana Avolio
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Giuseppe Quero
- Chirurgia Digestiva, Fondazione Policlinico Universitario Agostino Gemelli IRCCS Roma Italy
| | - Vincenzo Tondolo
- Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Enrico Celestino Nista
- Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | | | - Saverio Cinti
- Department of Clinical and Experimental Medicine Center of Obesity Università Politecnica delle Marche Ancona Italy
| | - Sergio Alfieri
- Chirurgia Digestiva, Fondazione Policlinico Universitario Agostino Gemelli IRCCS Roma Italy
| | - Antonio Gasbarrini
- Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Andrea Giaccari
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy.
| |
Collapse
|
3
|
Yang Y, Sun W, Yang F, Liang T, Li CL, Wang Y, Wang XL, Wang RR, Wu SC, Chen J. High energy diet-induced prediabetic neuropathic pain is mediated by reduction of SIRT6 negative control of both spinal and peripheral neuroinflammation. Neuroscience 2025; 569:58-66. [PMID: 39909339 DOI: 10.1016/j.neuroscience.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/16/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Prediabetic neuropathic pain has been classified as peripheral neuropathic pain associated with polyneuropathy caused by impaired glucose tolerance or impaired fasting glucose, which is a preclinical stage and might develop type 2 diabetes mellitus. Our previous research highlighted that prediabetes is accompanied by dramatic bilateral mechanical hyperalgesia following high energy diet (HED) which results in myelin and axonal degenerations along somatosensory system. However, the pathogenic mechanisms underlying prediabetic neuropathic pain remain unclear. The nuclear sirtuin 6 (SIRT6) is a crucial deacetylase in the regulation of multiple cellular biological processes, such as DNA repair, genome stability, inflammation and metabolic homeostasis. In current study, we show that the expressions of SIRT6 were significantly decreased, while its downstream NF-κB and proinflammatory mediator IL-6 and IL-1β were significantly increased in both dorsal root ganglia (DRG) and spinal dorsal horn of rats with prediabetic neuropathic pain induced by HED. Moreover, siRNA-SIRT6 treatment induced a significant reduction in bilateral paw withdrawal mechanical thresholds, indicating that SIRT6 down-regulation contributed to prediabetic neuropathic pain induced by HED. Furthermore, it was also found that SIRT6 reduction induced the activation of HMGB1 via disinhibition of NF-κB in both DRG and spinal dorsal horn of prediabetic rats. In conclusion, prediabetic neuropathic pain is caused by SIRT6 reduction through upregulating HMGB1-RAGE signaling at both peripheral and spinal levels.
Collapse
Affiliation(s)
- Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Fan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Ting Liang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China
| | - Shuang-Chan Wu
- Sanhang Institute for Brain Science and Technology, Northwestern Polytechnical University, Xi'an 710129 Shaanxi Province, PR China.
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038 Shaanxi Province, PR China; Sanhang Institute for Brain Science and Technology, Northwestern Polytechnical University, Xi'an 710129 Shaanxi Province, PR China.
| |
Collapse
|
4
|
Bonnefond A, Florez JC, Loos RJF, Froguel P. Dissection of type 2 diabetes: a genetic perspective. Lancet Diabetes Endocrinol 2025; 13:149-164. [PMID: 39818223 DOI: 10.1016/s2213-8587(24)00339-5] [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] [Received: 07/14/2024] [Revised: 09/11/2024] [Accepted: 10/30/2024] [Indexed: 01/18/2025]
Abstract
Diabetes is a leading cause of global mortality and disability, and its economic burden is substantial. This Review focuses on type 2 diabetes, which makes up 90-95% of all diabetes cases. Type 2 diabetes involves a progressive loss of insulin secretion often alongside insulin resistance and metabolic syndrome. Although obesity and a sedentary lifestyle are considerable contributors, research over the last 25 years has shown that type 2 diabetes develops on a predisposing genetic background, with family and twin studies indicating considerable heritability (ie, 31-72%). This Review explores type 2 diabetes from a genetic perspective, highlighting insights into its pathophysiology and the implications for precision medicine. More specifically, the traditional understanding of type 2 diabetes genetics has focused on a dichotomy between monogenic and polygenic forms. However, emerging evidence suggests a continuum that includes monogenic, oligogenic, and polygenic contributions, revealing their complementary roles in type 2 diabetes pathophysiology. Recent genetic studies provide deeper insights into disease mechanisms and pave the way for precision medicine approaches that could transform type 2 diabetes management. Additionally, the effect of environmental factors on type 2 diabetes, particularly from epigenetic modifications, adds another layer of complexity to understanding and addressing this multifaceted disease.
Collapse
Affiliation(s)
- Amélie Bonnefond
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France; Department of Metabolism, Imperial College London, London, UK.
| | - Jose C Florez
- Center for Genomic Medicine and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ruth J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philippe Froguel
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France; Department of Metabolism, Imperial College London, London, UK.
| |
Collapse
|
5
|
Pesta D, Anadol-Schmitz E, Sarabhai T, Op den Kamp Y, Gancheva S, Trinks N, Zaharia OP, Mastrototaro L, Lyu K, Habets I, Op den Kamp-Bruls YMH, Dewidar B, Weiss J, Schrauwen-Hinderling V, Zhang D, Gaspar RC, Strassburger K, Kupriyanova Y, Al-Hasani H, Szendroedi J, Schrauwen P, Phielix E, Shulman GI, Roden M. Determinants of increased muscle insulin sensitivity of exercise-trained versus sedentary normal weight and overweight individuals. SCIENCE ADVANCES 2025; 11:eadr8849. [PMID: 39742483 DOI: 10.1126/sciadv.adr8849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025]
Abstract
The athlete's paradox states that intramyocellular triglyceride accumulation associates with insulin resistance in sedentary but not in endurance-trained humans. Underlying mechanisms and the role of muscle lipid distribution and composition on glucose metabolism remain unclear. We compared highly trained athletes (ATHL) with sedentary normal weight (LEAN) and overweight-to-obese (OVWE) male and female individuals. This observational study found that ATHL show higher insulin sensitivity, muscle mitochondrial content, and capacity, but lower activation of novel protein kinase C (nPKC) isoforms, despite higher diacylglycerol concentrations. Notably, sedentary but insulin sensitive OVWE feature lower plasma membrane-to-mitochondria sn-1,2-diacylglycerol ratios. In ATHL, calpain-2, which cleaves nPKC, negatively associates with PKCε activation and positively with insulin sensitivity along with higher GLUT4 and hexokinase II content. These findings contribute to explaining the athletes' paradox by demonstrating lower nPKC activation, increased calpain, and mitochondrial partitioning of bioactive diacylglycerols, the latter further identifying an obesity subtype with increased insulin sensitivity (NCT03314714).
Collapse
Affiliation(s)
- Dominik Pesta
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Centre for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Evrim Anadol-Schmitz
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Theresia Sarabhai
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Yvo Op den Kamp
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Sofiya Gancheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Nina Trinks
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Oana-Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Kun Lyu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ivo Habets
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Yvonne M H Op den Kamp-Bruls
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Bedair Dewidar
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Jürgen Weiss
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Vera Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Klaus Strassburger
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology, Diabetology and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Schrauwen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- Leiden University Medical Center, Clinical Epidemiology, Leiden, Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
6
|
Onthoni DD, Chen YE, Lai YH, Li GH, Zhuang YS, Lin HM, Hsiao YP, Onthoni AI, Chiou HY, Chung RH. Clustering-based risk stratification of prediabetes populations: Insights from the Taiwan and UK Biobanks. J Diabetes Investig 2025; 16:25-35. [PMID: 39387466 DOI: 10.1111/jdi.14328] [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: 06/06/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024] Open
Abstract
AIMS/INTRODUCTION This study aimed to identify low- and high-risk diabetes groups within prediabetes populations using data from the Taiwan Biobank (TWB) and UK Biobank (UKB) through a clustering-based Unsupervised Learning (UL) approach, to inform targeted type 2 diabetes (T2D) interventions. MATERIALS AND METHODS Data from TWB and UKB, comprising clinical and genetic information, were analyzed. Prediabetes was defined by glucose thresholds, and incident T2D was identified through follow-up data. K-means clustering was performed on prediabetes participants using significant features determined through logistic regression and LASSO. Cluster stability was assessed using mean Jaccard similarity, silhouette score, and the elbow method. RESULTS We identified two stable clusters representing high- and low-risk diabetes groups in both biobanks. The high-risk clusters showed higher diabetes incidence, with 15.7% in TWB and 13.0% in UKB, compared to 7.3% and 9.1% in the low-risk clusters, respectively. Notably, males were predominant in the high-risk groups, constituting 76.6% in TWB and 52.7% in UKB. In TWB, the high-risk group also exhibited significantly higher BMI, fasting glucose, and triglycerides, while UKB showed marginal significance in BMI and other metabolic indicators. Current smoking was significantly associated with increased diabetes risk in the TWB high-risk group (P < 0.001). Kaplan-Meier curves indicated significant differences in diabetes complication incidences between clusters. CONCLUSIONS UL effectively identified risk-specific groups within prediabetes populations, with high-risk groups strongly associated male gender, higher BMI, smoking, and metabolic markers. Tailored preventive strategies, particularly for young males in Taiwan, are crucial to reducing T2D risk.
Collapse
Affiliation(s)
- Djeane Debora Onthoni
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Ying-Erh Chen
- Department of Risk Management and Insurance, Tamkang University, New Taipei City, Taiwan
| | - Yi-Hsuan Lai
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Guo-Hung Li
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Yong-Sheng Zhuang
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Hong-Ming Lin
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Yu-Ping Hsiao
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Ade Indra Onthoni
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Hung-Yi Chiou
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Ren-Hua Chung
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| |
Collapse
|
7
|
Blüher M. Understanding Adipose Tissue Dysfunction. J Obes Metab Syndr 2024; 33:275-288. [PMID: 39734091 PMCID: PMC11704217 DOI: 10.7570/jomes24013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/08/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
Diseases affecting adipose tissue (AT) function include obesity, lipodystrophy, and lipedema, among others. Both a lack of and excess AT are associated with increased risk for developing diseases including type 2 diabetes mellitus, hypertension, obstructive sleep apnea, and some types of cancer. However, individual risk of developing cardiometabolic and other 'obesity-related' diseases is not entirely determined by fat mass. Rather than excess fat accumulation, AT dysfunction may represent the mechanistic link between obesity and comorbid diseases. There are people who remain metabolically healthy despite obesity, whereas people with normal weight or very low subcutaneous AT mass may develop typically obesity-related diseases. AT dysfunction is characterized by adipocyte hypertrophy, impaired subcutaneous AT expandability (ectopic fat deposition), hypoxia, a variety of stress, inflammatory processes, and the release of proinflammatory, diabetogenic, and atherogenic signals. Genetic and environmental factors might contribute to AT heterogeneity either alone or via interaction with intrinsic biological factors. However, many questions remain regarding the mechanisms of AT dysfunction initiation and whether and how it could be reversed. Do AT signatures define clinically relevant subtypes of obesity? Is the cellular composition of AT associated with variation in obesity phenotypes? What roles do environmental compounds play in the manifestation of AT dysfunction? Answers to these and other questions may explain AT disease mechanisms and help to define strategies for improving AT health. This review focuses on recent advances in our understanding of AT biology.
Collapse
Affiliation(s)
- Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
8
|
Metwally AA, Perelman D, Park H, Wu Y, Jha A, Sharp S, Celli A, Ayhan E, Abbasi F, Gloyn AL, McLaughlin T, Snyder MP. Prediction of metabolic subphenotypes of type 2 diabetes via continuous glucose monitoring and machine learning. Nat Biomed Eng 2024:10.1038/s41551-024-01311-6. [PMID: 39715896 DOI: 10.1038/s41551-024-01311-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 11/01/2024] [Indexed: 12/25/2024]
Abstract
The classification of type 2 diabetes and prediabetes does not consider heterogeneity in the pathophysiology of glucose dysregulation. Here we show that prediabetes is characterized by metabolic heterogeneity, and that metabolic subphenotypes can be predicted by the shape of the glucose curve measured via a continuous glucose monitor (CGM) during standardized oral glucose-tolerance tests (OGTTs) performed in at-home settings. Gold-standard metabolic tests in 32 individuals with early glucose dysregulation revealed dominant or co-dominant subphenotypes (muscle or hepatic insulin-resistance phenotypes in 34% of the individuals, and β-cell-dysfunction or impaired-incretin-action phenotypes in 40% of them). Machine-learning models trained with glucose time series from OGTTs from the 32 individuals predicted the subphenotypes with areas under the curve (AUCs) of 95% for muscle insulin resistance, 89% for β-cell deficiency and 88% for impaired incretin action. With CGM-generated glucose curves obtained during at-home OGTTs, the models predicted the muscle-insulin-resistance and β-cell-deficiency subphenotypes of 29 individuals with AUCs of 88% and 84%, respectively. At-home identification of metabolic subphenotypes via a CGM may aid the risk stratification of individuals with early glucose dysregulation.
Collapse
Affiliation(s)
- Ahmed A Metwally
- Department of Genetics, Stanford University, Stanford, CA, USA
- Systems and Biomedical Engineering Department, Cairo University, Giza, Egypt
- Google LLC, Mountain View, CA, USA
| | - Dalia Perelman
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Heyjun Park
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yue Wu
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Alokkumar Jha
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Seth Sharp
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Ekrem Ayhan
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Fahim Abbasi
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Anna L Gloyn
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Centre, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
9
|
Franks PW, Sargent JL. Diabetes and obesity: leveraging heterogeneity for precision medicine. Eur Heart J 2024; 45:5146-5155. [PMID: 39523563 DOI: 10.1093/eurheartj/ehae746] [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: 06/01/2024] [Revised: 08/06/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
The increasing prevalence of diabetes, obesity, and their cardiometabolic sequelae present major global health challenges and highlight shortfalls of current approaches to the prevention and treatment of these conditions. Representing the largest global burden of morbidity and mortality, the pathobiological processes underlying cardiometabolic diseases are in principle preventable and, even when disease is manifest, sometimes reversable. Nevertheless, with current clinical and public health strategies, goals of widespread prevention and remission remain largely aspirational. Application of precision medicine approaches that reduce errors and improve accuracy in medical and health recommendations has potential to accelerate progress towards these goals. Precision medicine must also maintain safety and ideally be cost-effective, as well as being compatible with an individual's preferences, capabilities, and needs. Initial progress in precision medicine was made in the context of rare diseases, with much focus on pharmacogenetic studies, owing to the cause of these diseases often being attributable to highly penetrant single gene mutations. By contrast, most obesity and type 2 diabetes are heterogeneous in aetiology and clinical presentation, underpinned by complex interactions between genetic and non-genetic factors. The heterogeneity of these conditions can be leveraged for development of approaches for precision therapies. Adequate characterization of the heterogeneity in cardiometabolic disease necessitates diversity of and synthesis across data types and research methods, ideally culminating in precision trials and real-world application of precision medicine approaches. This State-of-the-Art Review provides an overview of the current state of the science of precision medicine, as well as outlining a roadmap for study designs that maximise opportunities and address challenges to clinical implementation of precision medicine approaches in obesity and diabetes.
Collapse
Affiliation(s)
- Paul W Franks
- Department of Clinical Sciences, Lund University, Helsingborg Hospital, Charlotte Yhlens gata 10, 251 87 Helsingborg, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jennifer L Sargent
- School of Public Health, Imperial College London, White City Campus, 80-92 Wood Lane, London, W12 0BZ, United Kingdom
- BabelFisk, Hälsovägen 9, Helsingborg, 252 21 Sweden
| |
Collapse
|
10
|
Vlaar APJ, Myatra SN, Jung C. Artificial intelligence to enhance hemodynamic management in the ICU. Intensive Care Med 2024:10.1007/s00134-024-07752-6. [PMID: 39714611 DOI: 10.1007/s00134-024-07752-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2024] [Indexed: 12/24/2024]
Affiliation(s)
- Alexander P J Vlaar
- Department of Intensive Care Medicine, Amsterdam UMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - Sheila N Myatra
- Department of Anaesthesiology, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine and Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
11
|
Stroebel BM, Gadgil M, Lewis K, Longoria K, Zhang L, Flowers E. Prediabetes Phenotype Clusters in the Diabetes Prevention Program Study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626435. [PMID: 39677703 PMCID: PMC11642785 DOI: 10.1101/2024.12.02.626435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Objective The purpose of this study was to apply clustering methods to identify and characterize prediabetes phenotypes and their relationships with treatment arm and type 2 diabetes (T2D) outcomes in the Diabetes Prevention Program (DPP), and to compare the utility of additional clustering measures in phenotype characterization and T2D risk stratification. Research Design and Methods This was a secondary analysis of data from a subset of participants (n=994) from the previously completed Diabetes Prevention Program trial. Unsupervised k-means clustering analysis was applied to derive the optimal number of clusters of participants based on common clinical risk factors alone or common risk factors plus more comprehensive measures of glucose tolerance and body composition. Results Five clusters were derived from common clinical characteristics and the addition of comprehensive measures of glucose tolerance and body composition. Within each modeling approach, participants show significantly different levels of risk factors. The clinical only model showed higher accuracy for time to T2D, however the more comprehensive models further differentiated a metabolically health overweight phenotype. For both models, the greatest differentiation in determining time to T2D was in the metformin arm of the trial. Conclusions Data driven clustering of patients with prediabetes allows for identification of prediabetes phenotypes at greater risk for disease progression and responses to risk reduction interventions. Further investigation into phenotypic differences in treatment response could enable better personalization of prediabetes and T2D prevention and treatment choices.
Collapse
|
12
|
Liu S, Zhu J, Zhong H, Wu C, Xue H, Darst BF, Guo X, Durda P, Tracy RP, Liu Y, Johnson WC, Taylor KD, Manichaikul AW, Goodarzi MO, Gerszten RE, Clish CB, Chen YDI, Highland H, Haiman CA, Gignoux CR, Lange L, Conti DV, Raffield LM, Wilkens L, Marchand LL, North KE, Young KL, Loos RJ, Buyske S, Matise T, Peters U, Kooperberg C, Reiner AP, Yu B, Boerwinkle E, Sun Q, Rooney MR, Echouffo-Tcheugui JB, Daviglus ML, Qi Q, Mancuso N, Li C, Deng Y, Manning A, Meigs JB, Rich SS, Rotter JI, Wu L. Identification of proteins associated with type 2 diabetes risk in diverse racial and ethnic populations. Diabetologia 2024; 67:2754-2770. [PMID: 39349773 DOI: 10.1007/s00125-024-06277-3] [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: 12/18/2023] [Accepted: 07/16/2024] [Indexed: 11/29/2024]
Abstract
AIMS/HYPOTHESIS Several studies have reported associations between specific proteins and type 2 diabetes risk in European populations. To better understand the role played by proteins in type 2 diabetes aetiology across diverse populations, we conducted a large proteome-wide association study using genetic instruments across four racial and ethnic groups: African; Asian; Hispanic/Latino; and European. METHODS Genome and plasma proteome data from the Multi-Ethnic Study of Atherosclerosis (MESA) study involving 182 African, 69 Asian, 284 Hispanic/Latino and 409 European individuals residing in the USA were used to establish protein prediction models by using potentially associated cis- and trans-SNPs. The models were applied to genome-wide association study summary statistics of 250,127 type 2 diabetes cases and 1,222,941 controls from different racial and ethnic populations. RESULTS We identified three, 44 and one protein associated with type 2 diabetes risk in Asian, European and Hispanic/Latino populations, respectively. Meta-analysis identified 40 proteins associated with type 2 diabetes risk across the populations, including well-established as well as novel proteins not yet implicated in type 2 diabetes development. CONCLUSIONS/INTERPRETATION Our study improves our understanding of the aetiology of type 2 diabetes in diverse populations. DATA AVAILABILITY The summary statistics of multi-ethnic type 2 diabetes GWAS of MVP, DIAMANTE, Biobank Japan and other studies are available from The database of Genotypes and Phenotypes (dbGaP) under accession number phs001672.v3.p1. MESA genetic, proteome and covariate data can be accessed through dbGaP under phs000209.v13.p3. All code is available on GitHub ( https://github.com/Arthur1021/MESA-1K-PWAS ).
Collapse
Affiliation(s)
- Shuai Liu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Jingjing Zhu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haoran Xue
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Burcu F Darst
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peter Durda
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - Russell P Tracy
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - W Craig Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ani W Manichaikul
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Heather Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher R Gignoux
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Leslie Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Laura M Raffield
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lynne Wilkens
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Loïc Le Marchand
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ruth J Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steve Buyske
- Department of Statistics, Rutgers University, Piscataway, NJ, USA
| | - Tara Matise
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Justin B Echouffo-Tcheugui
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nicholas Mancuso
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Alisa Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA.
| |
Collapse
|
13
|
Niu Y, Wang S, Gao P, Ren X, Li F, Liu Z, Wang L, Peng H, Ju S. Photo-transformation of biochar-derived dissolved organic matter and its binding with phenanthrene/9-phenanthrol: The role of functional group and pyrolysis temperature. BIORESOURCE TECHNOLOGY 2024; 413:131547. [PMID: 39343176 DOI: 10.1016/j.biortech.2024.131547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/22/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
This study explores the physicochemical attributes of dissolved organic matter from rice straw biochar (BDOM) at varying pyrolysis temperatures and photo-irradiation conditions, focusing on the binding mechanisms of phenanthrene (PHE) and 9-phenanthrol (PTR) using multiple spectroscopic techniques and fluorescence quenching. Following 20 h of photo-irradiation, only 11.3 % of BDOM underwent mineralization, forming new CH3/CH2/CH aliphatics structures. BDOM from biochar produced by pyrolysis at 400°C exhibited a stronger binding affinity with PHE and PTR, achieving 44 % and 52 % maximum binding, respectively. Static and dynamic quenching governed PHE and PTR binding, which was influenced by temperature. Photo-irradiated BDOM showed enhanced binding with PHE, attributed to increased aliphatic content. Hydrogen bond and π-π electron-donor-acceptor (EDA) interactions dominated PTR binding, while π-π interactions and hydrophobic interactions controlled PHE. This study provides valuable insights into BDOM photochemical behaviors and their impact on the environmental fate of polycyclic aromatic hydrocarbons (PAHs) after BDOM photo-irradiation.
Collapse
Affiliation(s)
- Yifan Niu
- Faculty of Modern Agricultural Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China; Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650093, China
| | - Siyao Wang
- Faculty of Modern Agricultural Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Peng Gao
- City College, Kunming University of Science & Technology, Kunming, Yunnan 650051, China
| | - Xin Ren
- Faculty of Modern Agricultural Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Fangfang Li
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Zhanpeng Liu
- Faculty of Modern Agricultural Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Lin Wang
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Hongbo Peng
- Faculty of Modern Agricultural Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China; Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China.
| | - Shaohua Ju
- Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650093, China.
| |
Collapse
|
14
|
Kanasaki K, Ueki K, Nangaku M. Diabetic kidney disease: the kidney disease relevant to individuals with diabetes. Clin Exp Nephrol 2024; 28:1213-1220. [PMID: 39031296 PMCID: PMC11621156 DOI: 10.1007/s10157-024-02537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/04/2024] [Indexed: 07/22/2024]
Abstract
In individuals with diabetes, chronic kidney disease (CKD) is a major comorbidity. However, it appears that there is worldwide confusion regarding which term should be used to describe CKD complicated with diabetes: diabetic nephropathy, diabetic kidney disease (DKD), CKD with diabetes, diabetes and CKD, etc. Similar confusion has also been reported in Japan. Therefore, to provide clarification, the Japanese Diabetes Society and the Japanese Society of Nephrology collaborated to update the corresponding Japanese term to describe DKD and clearly define the concept of DKD. In this review, we briefly described the history of kidney complications in individuals with diabetes and the Japanese definition of the DKD concept and provided our rationale for these changes.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Department of Internal Medicine 1, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo, 693-8501, Japan.
- The Center for Integrated Kidney Research and Advance, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo, 693-8501, Japan.
| | - Kohjiro Ueki
- Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Herder C, Rizzo M, Roden M. Precision diabetology: Where do we stand now? J Diabetes Complications 2024; 38:108899. [PMID: 39477695 DOI: 10.1016/j.jdiacomp.2024.108899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024]
Affiliation(s)
- Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Manfredi Rizzo
- Unit of Diabetes and Cardiometabolic Prevention, University Hospital of Palermo, Palermo, Italy; School of Medicine, Department of Health Promotion, Mother and Child Care Internal Medicine and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - 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 (DZD), München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
16
|
Choi W, Park M, Park S, Park JY, Hong AR, Yoon JH, Ha KH, Kim DJ, Kim HK, Kang HC. Combined impact of prediabetes and hepatic steatosis on cardiometabolic outcomes in young adults. Cardiovasc Diabetol 2024; 23:422. [PMID: 39574105 PMCID: PMC11583572 DOI: 10.1186/s12933-024-02516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024] Open
Abstract
OBJECTIVES This study aimed to investigate the impact of hepatic steatosis on cardiometabolic outcomes in young adults with prediabetes. METHODS A nationwide cohort study was conducted with 896,585 young adults under 40 years old without diabetes or previous history of cardiovascular disease. Hepatic steatosis was identified using a fatty liver index of ≥ 60. The outcomes of this study were incident diabetes (DM) and composite major adverse cardiovascular events (MACE), including myocardial infarction, stroke, or cardiovascular death. RESULTS During a median follow-up of 11.8 years, 27,437 (3.1%) incident DM cases and 6,584 (0.7%) MACE cases were recorded. Young adults with prediabetes had a significantly higher risk of incident DM (hazard ratio [HR]: 2.81; 95% confidence interval [CI]: 2.74-2.88; P-value: <0.001) and composite MACE risk (HR: 1.10; 95% CI: 1.03-1.17; P-value: 0.003) compared to individuals with normoglycemia, after adjusting for relevant covariates. Stratification based on hepatic steatosis showed that the combination of prediabetes and hepatic steatosis posed the highest risk for these outcomes, after adjusting for relevant covariates. For incident DM, the HRs (95% CI; P-value) were: 3.15 (3.05-3.26; <0.001) for prediabetes without hepatic steatosis, 2.89 (2.78-3.01; <0.001) for normoglycemia with hepatic steatosis, and 6.60 (6.33-6.87; <0.001) for prediabetes with hepatic steatosis. For composite MACE, the HRs (95% CI; P-value) were 1.05 (0.97-1.13; 0.235) for prediabetes without hepatic steatosis, 1.39 (1.27-1.51; <0.001) for normoglycemia with hepatic steatosis, and 1.60 (1.44-1.78; <0.001) for prediabetes with hepatic steatosis. CONCLUSIONS Prediabetes and hepatic steatosis additively increased the risk of cardiometabolic outcomes in young adults. These findings hold significance for physicians as they provide insights into assessing high-risk individuals among young adults with prediabetes.
Collapse
Affiliation(s)
- Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea.
- Department of Biological Chemistry, University of California Irvine School of Medicine, Irvine, CA, USA.
| | - Minae Park
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Sojeong Park
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Ji Yong Park
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - A Ram Hong
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - Jee Hee Yoon
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - Kyoung Hwa Ha
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea.
| | - Ho-Cheol Kang
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| |
Collapse
|
17
|
Wang J, Gao B, Wang J, Liu W, Yuan W, Chai Y, Ma J, Ma Y, Kong G, Liu M. Identifying subtypes of type 2 diabetes mellitus based on real-world electronic medical record data in China. Diabetes Res Clin Pract 2024; 217:111872. [PMID: 39332534 DOI: 10.1016/j.diabres.2024.111872] [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: 06/21/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
AIMS To replicate the European subtypes of type 2 diabetes mellitus (T2DM) in the Chinese diabetes population and investigate the risk of complications in different subtypes. METHODS A diabetes cohort using real-world patient data was constructed, and clustering was employed to subgroup the T2DM patients. Kaplan-Meier analysis and the Cox models were used to analyze the association between diabetes subtypes and the risk of complications. RESULTS A total of 2,652 T2DM patients with complete clustering data were extracted. Among them, 466 (17.57 %) were classified as severe insulin-deficient diabetes (SIDD), 502 (18.93 %) as severe insulin-resistant diabetes (SIRD), 672 (25.34 %) as mild obesity-related diabetes (MOD), and 1,012 (38.16 %) as mild age-related diabetes (MARD). The risk of chronic kidney disease (CKD) and diabetic retinopathy (DR) were different in the four subtypes. Compared with MARD, SIRD had a higher risk of CKD (HR 2.40 [1.16, 4.96]), and SIDD had a higher risk of DR (HR 2.16 [1.11, 4.20]). The risk of stroke and coronary events had no difference. CONCLUSIONS The European T2DM subtypes can be replicated in the Chinese diabetes population. The risk of CKD and DR varied among different subtypes, indicating that proper interventions can be taken to prevent specific complications in different subtypes.
Collapse
Affiliation(s)
- Jiayu Wang
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Bixia Gao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Jinwei Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Wenwen Liu
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Weijia Yuan
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Yangfan Chai
- Peking University Chongqing Research Institute of Big Data, Chongqing 100871, China
| | - Jun Ma
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Yangyang Ma
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Guilan Kong
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China.
| | - Minchao Liu
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
18
|
Kanasaki K, Nangaku M, Ueki K. 'DKD' as the kidney disease relevant to individuals with diabetes. Diabetol Int 2024; 15:673-676. [PMID: 39469547 PMCID: PMC11512933 DOI: 10.1007/s13340-024-00747-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Indexed: 10/30/2024]
Abstract
Even though chronic kidney disease (CKD) is a significant comorbidity in individuals with diabetes, there appears to be worldwide confusion regarding the terminology used to describe it, including diabetic nephropathy, diabetic kidney disease (DKD), CKD with diabetes, diabetes and CKD, etc. In Japan, we have encountered similar confusion regarding the terminology used to describe kidney disease in individuals with diabetes, especially when written in Japanese due to terminological similarities in Chinese characters. The primary issue in Japan was deciphering the significance of "Diabetic," specifically whether it is an essential attribute of the condition itself. The confusions may arise from the deficiencies in establishing a clear criterion for the disease concept, whether it is diabetic nephropathy or DKD. Furthermore, among specialists in the field, each may have their own concept of the disease. In this regard, the Japanese Diabetes Society and the Japanese Society of Nephrology updated the corresponding Japanese term for DKD and defined the concept of DKD with rationale. The goal of these efforts should be the future improvement of the prognosis of DKD patients, the stakeholders.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Department of Internal Medicine 1, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo, 693-8501 Japan
- The Center for Integrated Kidney Research and Advance, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo, 693-8501 Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kohjiro Ueki
- Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Metwally AA, Perelman D, Park H, Wu Y, Jha A, Sharp S, Celli A, Ayhan E, Abbasi F, Gloyn AL, McLaughlin T, Snyder M. Predicting Type 2 Diabetes Metabolic Phenotypes Using Continuous Glucose Monitoring and a Machine Learning Framework. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.20.24310737. [PMID: 39108516 PMCID: PMC11302614 DOI: 10.1101/2024.07.20.24310737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Type 2 diabetes (T2D) and prediabetes are classically defined by the level of fasting glucose or surrogates such as hemoglobin HbA1c. This classification does not take into account the heterogeneity in the pathophysiology of glucose dysregulation, the identification of which could inform targeted approaches to diabetes treatment and prevention and/or predict clinical outcomes. We performed gold-standard metabolic tests in a cohort of individuals with early glucose dysregulation and quantified four distinct metabolic subphenotypes known to contribute to glucose dysregulation and T2D: muscle insulin resistance, β-cell dysfunction, impaired incretin action, and hepatic insulin resistance. We revealed substantial inter-individual heterogeneity, with 34% of individuals exhibiting dominance or co-dominance in muscle and/or liver IR, and 40% exhibiting dominance or co-dominance in β-cell and/or incretin deficiency. Further, with a frequently-sampled oral glucose tolerance test (OGTT), we developed a novel machine learning framework to predict metabolic subphenotypes using features from the dynamic patterns of the glucose time-series ("shape of the glucose curve"). The glucose time-series features identified insulin resistance, β-cell deficiency, and incretin defect with auROCs of 95%, 89%, and 88%, respectively. These figures are superior to currently-used estimates. The prediction of muscle insulin resistance and β-cell deficiency were validated using an independent cohort. We then tested the ability of glucose curves generated by a continuous glucose monitor (CGM) worn during at-home OGTTs to predict insulin resistance and β-cell deficiency, yielding auROC of 88% and 84%, respectively. We thus demonstrate that the prediabetic state is characterized by metabolic heterogeneity, which can be defined by the shape of the glucose curve during standardized OGTT, performed in a clinical research unit or at-home setting using CGM. The use of at-home CGM to identify muscle insulin resistance and β-cell deficiency constitutes a practical and scalable method by which to risk stratify individuals with early glucose dysregulation and inform targeted treatment to prevent T2D.
Collapse
Affiliation(s)
- Ahmed A. Metwally
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Dalia Perelman
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Heyjun Park
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Yue Wu
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Alokkumar Jha
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Seth Sharp
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Alessandra Celli
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Ekrem Ayhan
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Fahim Abbasi
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Anna L Gloyn
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
- Stanford Diabetes Research Centre, Stanford University, Stanford, CA 94305, USA
| | - Tracey McLaughlin
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Michael Snyder
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| |
Collapse
|
20
|
Dietrich JW, Abood A, Dasgupta R, Anoop S, Jebasingh FK, Spurgeon R, Thomas N, Boehm BO. A novel simple disposition index (SPINA-DI) from fasting insulin and glucose concentration as a robust measure of carbohydrate homeostasis. J Diabetes 2024; 16:e13525. [PMID: 38169110 PMCID: PMC11418405 DOI: 10.1111/1753-0407.13525] [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: 06/26/2023] [Revised: 11/17/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
AIMS The widely used dynamic disposition index, derived from oral glucose tolerance testing, is an integrative measure of the homeostatic performance of the insulin-glucose feedback control. Its collection is, however, time consuming and expensive. We, therefore, pursued the question if such a measure can be calculated at baseline/fasting conditions using plasma concentrations of insulin and glucose. METHODS A new fasting-based disposition index (structure parameter inference approach-disposition index [SPINA-DI]) was calculated as the product of the reconstructed insulin receptor gain (SPINA-GR) times the secretory capacity of pancreatic beta cells (SPINA-GBeta). The novel index was evaluated in computer simulations and in three independent, multiethnic cohorts. The objectives were distribution in various populations, diagnostic performance, reliability and correlation to established physiological biomarkers of carbohydrate metabolism. RESULTS Mathematical and in-silico analysis demonstrated SPINA-DI to mirror the hyperbolic relationship between insulin sensitivity and beta-cell function and to represent an optimum of the homeostatic control. It significantly correlates to the oral glucose tolerance test based disposition index and other important physiological parameters. Furthermore, it revealed higher discriminatory power for the diagnosis of (pre)diabetes and superior retest reliability than other static and dynamic function tests of glucose homeostasis. CONCLUSIONS SPINA-DI is a novel simple reliable and inexpensive marker of insulin-glucose homeostasis suitable for screening purposes and a wider clinical application.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef HospitalRuhr University BochumBochumGermany
- Diabetes Centre Bochum/Hattingen, St. Elisabeth‐Hospital BlankensteinHattingenGermany
- Centre for Rare Endocrine Diseases, Ruhr Centre for Rare Diseases (CeSER)Ruhr University Bochum and Witten/Herdecke UniversityBochumGermany
- Centre for Diabetes TechnologyCatholic Hospitals BochumBochumGermany
| | - Assjana Abood
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef HospitalRuhr University BochumBochumGermany
- Diabetes Centre Bochum/Hattingen, St. Elisabeth‐Hospital BlankensteinHattingenGermany
- Centre for Rare Endocrine Diseases, Ruhr Centre for Rare Diseases (CeSER)Ruhr University Bochum and Witten/Herdecke UniversityBochumGermany
- Centre for Diabetes TechnologyCatholic Hospitals BochumBochumGermany
| | - Riddhi Dasgupta
- Department of Endocrinology, Diabetes and MetabolismChristian Medical CollegeVelloreIndia
| | - Shajith Anoop
- Department of Endocrinology, Diabetes and MetabolismChristian Medical CollegeVelloreIndia
| | - Felix K. Jebasingh
- Department of Endocrinology, Diabetes and MetabolismChristian Medical CollegeVelloreIndia
| | - R. Spurgeon
- Department of EndocrinologyBangalore Baptist HospitalBangaloreIndia
| | - Nihal Thomas
- Department of Endocrinology, Diabetes and MetabolismChristian Medical CollegeVelloreIndia
| | - Bernhard O. Boehm
- Lee Kong Chian School of MedicineNanyang Technological University SingaporeSingaporeSingapore
- King's College LondonSchool of Life Course & Population SciencesLondonUK
| |
Collapse
|
21
|
Tacke F, Horn P, Wai-Sun Wong V, Ratziu V, Bugianesi E, Francque S, Zelber-Sagi S, Valenti L, Roden M, Schick F, Yki-Järvinen H, Gastaldelli A, Vettor R, Frühbeck G, Dicker D. EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol 2024; 81:492-542. [PMID: 38851997 DOI: 10.1016/j.jhep.2024.04.031] [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] [Received: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 06/10/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
22
|
Hörber S, Prystupa K, Jacoby J, Fritsche A, Kleber ME, Moissl AP, Hellstern P, Peter A, März W, Wagner R, Heni M. Blood coagulation in Prediabetes clusters-impact on all-cause mortality in individuals undergoing coronary angiography. Cardiovasc Diabetol 2024; 23:306. [PMID: 39175055 PMCID: PMC11342575 DOI: 10.1186/s12933-024-02402-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Metabolic clusters can stratify subgroups of individuals at risk for type 2 diabetes mellitus and related complications. Since obesity and insulin resistance are closely linked to alterations in hemostasis, we investigated the association between plasmatic coagulation and metabolic clusters including the impact on survival. METHODS Utilizing data from the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, we assigned 917 participants without diabetes to prediabetes clusters, using oGTT-derived glucose and insulin, high-density lipoprotein cholesterol, triglycerides, and anthropometric data. We performed a comprehensive analysis of plasmatic coagulation parameters and analyzed their associations with mortality using proportional hazards models. Mediation analysis was performed to assess the effect of coagulation factors on all-cause mortality in prediabetes clusters. RESULTS Prediabetes clusters were assigned using published tools, and grouped into low-risk (clusters 1,2,4; n = 643) and high-risk (clusters 3,5,6; n = 274) clusters. Individuals in the high-risk clusters had a significantly increased risk of death (HR = 1.30; CI: 1.01 to 1.67) and showed significantly elevated levels of procoagulant factors (fibrinogen, FVII/VIII/IX), D-dimers, von-Willebrand factor, and PAI-1, compared to individuals in the low-risk clusters. In proportional hazards models adjusted for relevant confounders, elevated levels of fibrinogen, D-dimers, FVIII, and vWF were found to be associated with an increased risk of death. Multiple mediation analysis indicated that vWF significantly mediates the cluster-specific risk of death. CONCLUSIONS High-risk prediabetes clusters are associated with prothrombotic changes in the coagulation system that likely contribute to the increased mortality in those individuals at cardiometabolic risk. The hypercoagulable state observed in the high-risk clusters indicates an increased risk for cardiovascular and thrombotic diseases that should be considered in future risk stratification and therapeutic strategies.
Collapse
Affiliation(s)
- Sebastian Hörber
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany.
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| | - Katsiaryna Prystupa
- German Center for Diabetes Research, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Johann Jacoby
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Department for Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Angela P Moissl
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Hellstern
- Center of Hemostasis and Thrombosis Zurich, Zurich, Switzerland
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Robert Wagner
- German Center for Diabetes Research, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Martin Heni
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
23
|
Gómez-Peralta F, Pinés-Corrales PJ, Santos E, Cuesta M, González-Albarrán O, Azriel S. Diabetes Management Based on the Phenotype and Stage of the Disease: An Expert Proposal from the AGORA Diabetes Collaborative Group. J Clin Med 2024; 13:4839. [PMID: 39200982 PMCID: PMC11355114 DOI: 10.3390/jcm13164839] [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: 07/04/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Diabetes is a complex and rapidly growing disease with heterogeneous clinical presentations. Recent advances in molecular and genetic technologies have led to the identification of various subtypes of diabetes. These advancements offer the potential for a more precise, individualized approach to treatment, known as precision medicine. Recognizing high-risk phenotypes and intervening early and intensively is crucial. A staging system for type 1 diabetes has been proposed and accepted globally. In this article, we will explore the different methods for categorizing and classifying type 2 diabetes (T2D) based on clinical characteristics, progression patterns, risk of complications, and the use of molecular techniques for patient grouping. We, as a team of experts, will also present an easy-to-follow treatment plan and guidance for non-specialists, particularly primary care physicians, that integrates the classification and staging of diabetes. This will help ensure that the most suitable therapy is applied to the different types of T2D at each stage of the disease's progression.
Collapse
Affiliation(s)
| | - Pedro J. Pinés-Corrales
- Endocrinology and Nutrition Service, Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain;
| | - Estefanía Santos
- Endocrinology and Nutrition Service, Complejo Hospitalario de Burgos, 09006 Burgos, Spain;
| | - Martín Cuesta
- Endocrinology and Nutrition Service, Hospital Clínico San Carlos, 28040 Madrid, Spain;
| | | | - Sharona Azriel
- Endocrinology and Nutrition Service, Hospital Universitario Infanta Sofía, 28702 San Sebastián De Los Reyes, Spain;
| | | |
Collapse
|
24
|
Baars DP, Fondevila MF, Meijnikman AS, Nieuwdorp M. The central role of the gut microbiota in the pathophysiology and management of type 2 diabetes. Cell Host Microbe 2024; 32:1280-1300. [PMID: 39146799 DOI: 10.1016/j.chom.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024]
Abstract
The inhabitants of our intestines, collectively called the gut microbiome, comprise fungi, viruses, and bacterial strains. These microorganisms are involved in the fermentation of dietary compounds and the regulation of our adaptive and innate immune systems. Less known is the reciprocal interaction between the gut microbiota and type 2 diabetes mellitus (T2DM), as well as their role in modifying therapies to reduce associated morbidity and mortality. In this review, we aim to discuss the existing literature on gut microbial strains and their diet-derived metabolites involved in T2DM. We also explore the potential diagnostics and therapeutic avenues the gut microbiota presents for targeted T2DM management. Personalized treatment plans, driven by diet and medication based on the patient's microbiome and clinical markers, could optimize therapy.
Collapse
Affiliation(s)
- Daniel P Baars
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Marcos F Fondevila
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Abraham S Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands; Diabetes Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Gijbels A, Jardon KM, Trouwborst I, Manusama KC, Goossens GH, Blaak EE, Feskens EJ, Afman LA. Fasting and postprandial plasma metabolite responses to a 12-wk dietary intervention in tissue-specific insulin resistance: a secondary analysis of the PERSonalized glucose Optimization through Nutritional intervention (PERSON) randomized trial. Am J Clin Nutr 2024; 120:347-359. [PMID: 38851634 DOI: 10.1016/j.ajcnut.2024.05.027] [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: 02/22/2024] [Revised: 05/06/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND We previously showed that dietary intervention effects on cardiometabolic health were driven by tissue-specific insulin resistance (IR) phenotype: individuals with predominant muscle IR (MIR) benefited more from a low-fat, high-protein, and high-fiber (LFHP) diet, whereas individuals with predominant liver insulin resistance (LIR) benefited more from a high-monounsaturated fatty acid (HMUFA) diet. OBJECTIVES To further characterize the effects of LFHP and HMUFA diets and their interaction with tissue-specific IR, we investigated dietary intervention effects on fasting and postprandial plasma metabolite profile. METHODS Adults with MIR or LIR (40-75 y, BMI 25-40 kg/m2) were randomly assigned to a 12-wk HMUFA or LFHP diet (n = 242). After the exclusion of statin use, 214 participants were included in this prespecified secondary analysis. Plasma samples were collected before (T = 0) and after (T = 30, 60, 120, and 240 min) a high-fat mixed meal for quantification of 247 metabolite measures using nuclear magnetic resonance spectroscopy. RESULTS A larger reduction in fasting VLDL-triacylglycerol (TAG) and VLDL particle size was observed in individuals with MIR following the LFHP diet and those with LIR following the HMUFA diet, although no longer statistically significant after false discovery rate (FDR) adjustment. No IR phenotype-by-diet interactions were found for postprandial plasma metabolites assessed as total area under the curve (tAUC). Irrespective of IR phenotype, the LFHP diet induced greater reductions in postprandial plasma tAUC of the larger VLDL particles and small HDL particles, and TAG content in most VLDL subclasses and the smaller LDL and HDL subclasses (for example, VLDL-TAG tAUC standardized mean change [95% CI] LFHP = -0.29 [-0.43, -0.16] compared with HMUFA = -0.04 [-0.16, 0.09]; FDR-adjusted P for diet × time = 0.041). CONCLUSIONS Diet effects on plasma metabolite profiles were more pronounced than phenotype-by-diet interactions. An LFHP diet may be more effective than an HMUFA diet for reducing cardiometabolic risk in individuals with tissue-specific IR, irrespective of IR phenotype. Am J Clin Nutr 20xx;x:xx. This trial was registered at the clinicaltrials.gov registration (https://clinicaltrials.gov/study/NCT03708419?term=NCT03708419&rank=1) as NCT03708419 and CCMO registration (https://www.toetsingonline.nl/to/ccmo_search.nsf/fABRpop?readform&unids=3969AABCD9BA27FEC12587F1001BCC65) as NL63768.068.17.
Collapse
Affiliation(s)
- Anouk Gijbels
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands; Top Institute Food and Nutrition (TiFN), Wageningen, The Netherlands.
| | - Kelly M Jardon
- Top Institute Food and Nutrition (TiFN), Wageningen, The Netherlands; Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Inez Trouwborst
- Top Institute Food and Nutrition (TiFN), Wageningen, The Netherlands; Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Koen Cm Manusama
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E Blaak
- Top Institute Food and Nutrition (TiFN), Wageningen, The Netherlands; Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Edith Jm Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
26
|
Birkenfeld AL, Mohan V. Prediabetes remission for type 2 diabetes mellitus prevention. Nat Rev Endocrinol 2024; 20:441-442. [PMID: 38806698 DOI: 10.1038/s41574-024-00996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Affiliation(s)
- Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Department of Internal Medicine IV, Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
- Department of Diabetes, Life Sciences & Medicine Cardiovascular Medicine & Sciences, King's College London, London, UK.
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation, Chennai, India
- Dr. Mohan's Diabetes Specialities Centre, Chennai, India
| |
Collapse
|
27
|
Renklint R, Chninou Y, Heni M, Fritsche A, Haering HU, Wagner R, Otten J. Surrogate measures of first-phase insulin secretion versus reference methods intravenous glucose tolerance test and hyperglycemic clamp: a systematic review and meta-analyses. BMJ Open Diabetes Res Care 2024; 12:e004256. [PMID: 39013634 PMCID: PMC11268049 DOI: 10.1136/bmjdrc-2024-004256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024] Open
Abstract
INTRODUCTION In this systematic review, we investigated the diagnostic accuracy of surrogate measures of insulin secretion based on fasting samples and the oral glucose tolerance test (OGTT). The first phase of insulin secretion was calculated using two gold standard methods; the hyperglycemic clamp (HGC) test and intravenous glucose tolerance test (IVGTT). RESEARCH DESIGN AND METHODS We conducted searches in the PubMed, Cochrane Central, and Web of Science databases, the last of which was conducted at the end of June 2021. Studies were included that measured first-phase insulin secretion in adults using both a gold-standard reference method (either HGC or IVGTT) and one or more surrogate measures from either fasting samples, OGTT or a meal-tolerance test. QUADAS-2, a revised tool for the quality assessment of diagnostic accuracy studies, was used for quality assessment. Random-effects meta-analyses were performed to examine the correlation between first-phase measured with gold standard and surrogate methods. RESULTS A total of 33 articles, encompassing 5362 individuals with normal glucose tolerance, pre-diabetes or type 2 diabetes, were included in our systematic review. Homeostatic model assessment (HOMA)-beta and Insulinogenic Index 30 (IGI(30)) were the surrogate measures validated in the largest number of studies (17 and 13, respectively). HOMA-beta's pooled correlation to the reference methods was 0.48 (95% CI 0.40 to 0.56) The pooled correlation of IGI to the reference methods was 0.61 (95% CI 0.54 to 0.68). The surrogate measures with the highest correlation to the reference methods were Kadowaki (0.67 (95% CI 0.61 to 0.73)) and Stumvoll's first-phase secretion (0.65 (95% CI 0.58 to 0.71)), both calculated from an OGTT. CONCLUSIONS Surrogate measures from the first 30 min of an OGTT capture the first phase of insulin secretion and are a good choice for epidemiological studies. HOMA-beta has a moderate correlation to the reference methods but is not a measure of the first phase specifically. PROSPERO REGISTRATION NUMBER The meta-analysis was registered at PROSPERO (Id: CRD42020169064) before inclusion started.
Collapse
Affiliation(s)
- Rebecka Renklint
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| | - Youssef Chninou
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| | - Martin Heni
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital of Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Hans-Ulrich Haering
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, German Diabetes Center, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Julia Otten
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| |
Collapse
|
28
|
Heni M. The insulin resistant brain: impact on whole-body metabolism and body fat distribution. Diabetologia 2024; 67:1181-1191. [PMID: 38363340 PMCID: PMC11153284 DOI: 10.1007/s00125-024-06104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 12/19/2023] [Indexed: 02/17/2024]
Abstract
Insulin exerts its actions not only on peripheral organs but is also transported into the brain where it performs distinct functions in various brain regions. This review highlights recent advancements in our understanding of insulin's actions within the brain, with a specific emphasis on investigations in humans. It summarises current knowledge on the transport of insulin into the brain. Subsequently, it showcases robust evidence demonstrating the existence and physiological consequences of brain insulin action, while also introducing the presence of brain insulin resistance in humans. This pathophysiological condition goes along with an impaired acute modulation of peripheral metabolism in response to brain insulin action, particularly in the postprandial state. Furthermore, brain insulin resistance has been associated with long-term adiposity and an unfavourable adipose tissue distribution, thus implicating it in the pathogenesis of subgroups of obesity and (pre)diabetes that are characterised by distinct patterns of body fat distribution. Encouragingly, emerging evidence suggests that brain insulin resistance could represent a treatable entity, thereby opening up novel therapeutic avenues to improve systemic metabolism and enhance brain functions, including cognition. The review closes with an outlook towards prospective research directions aimed at further elucidating the clinical implications of brain insulin resistance. It emphasises the critical need to establish feasible diagnostic measures and effective therapeutic interventions.
Collapse
Affiliation(s)
- Martin Heni
- Division of Endocrinology and Diabetology, Department of Internal Medicine 1, University Hospital Ulm, Ulm, Germany.
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital of Tübingen, Tübingen, Germany.
| |
Collapse
|
29
|
Somasundaram A, Wu M, Reik A, Rupp S, Han J, Naebauer S, Junker D, Patzelt L, Wiechert M, Zhao Y, Rueckert D, Hauner H, Holzapfel C, Karampinos DC. Evaluating Sex-specific Differences in Abdominal Fat Volume and Proton Density Fat Fraction at MRI Using Automated nnU-Net-based Segmentation. Radiol Artif Intell 2024; 6:e230471. [PMID: 38809148 PMCID: PMC11294970 DOI: 10.1148/ryai.230471] [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: 10/27/2023] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024]
Abstract
Sex-specific abdominal organ volume and proton density fat fraction (PDFF) in people with obesity during a weight loss intervention was assessed with automated multiorgan segmentation of quantitative water-fat MRI. An nnU-Net architecture was employed for automatic segmentation of abdominal organs, including visceral and subcutaneous adipose tissue, liver, and psoas and erector spinae muscle, based on quantitative chemical shift-encoded MRI and using ground truth labels generated from participants of the Lifestyle Intervention (LION) study. Each organ's volume and fat content were examined in 127 participants (73 female and 54 male participants; body mass index, 30-39.9 kg/m2) and in 81 (54 female and 32 male participants) of these participants after an 8-week formula-based low-calorie diet. Dice scores ranging from 0.91 to 0.97 were achieved for the automatic segmentation. PDFF was found to be lower in visceral adipose tissue compared with subcutaneous adipose tissue in both male and female participants. Before intervention, female participants exhibited higher PDFF in subcutaneous adipose tissue (90.6% vs 89.7%; P < .001) and lower PDFF in liver (8.6% vs 13.3%; P < .001) and visceral adipose tissue (76.4% vs 81.3%; P < .001) compared with male participants. This relation persisted after intervention. As a response to caloric restriction, male participants lost significantly more visceral adipose tissue volume (1.76 L vs 0.91 L; P < .001) and showed a higher decrease in subcutaneous adipose tissue PDFF (2.7% vs 1.5%; P < .001) than female participants. Automated body composition analysis on quantitative water-fat MRI data provides new insights for understanding sex-specific metabolic response to caloric restriction and weight loss in people with obesity. Keywords: Obesity, Chemical Shift-encoded MRI, Abdominal Fat Volume, Proton Density Fat Fraction, nnU-Net ClinicalTrials.gov registration no. NCT04023942 Supplemental material is available for this article. Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
| | | | - Anna Reik
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Selina Rupp
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Jessie Han
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Stella Naebauer
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Daniela Junker
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Lisa Patzelt
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Meike Wiechert
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Yu Zhao
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Daniel Rueckert
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Hans Hauner
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Christina Holzapfel
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| | - Dimitrios C. Karampinos
- From the Department of Diagnostic and Interventional Radiology,
Klinikum rechts der Isar (A.S., M. Wu, S.R., J.H., S.N., D.J., L.P., D.C.K.),
Institute of Nutritional Medicine, School of Medicine (A.R., M. Wiechert, H.H.,
C.H.), TUM School of Computation, Information, and Technology (Y.Z., D.R.), TUM
School of Medicine and Health (D.R.), and Else Kröner Fresenius Center
for Nutritional Medicine, School of Medicine (H.H.), Technical University of
Munich, Ismaninger Str 22, 81675 Munich, Germany; Department of Computing,
Imperial College London, London, UK (D.R.); Department of Nutritional, Food and
Consumer Sciences, Fulda University of Applied Sciences, Fulda, Germany (C.H.);
and Munich Institute of Biomedical Engineering and Munich Data Science
Institute, Technical University of Munich, Garching, Germany (D.C.K.)
| |
Collapse
|
30
|
Li Y, Chen GC, Moon JY, Arthur R, Sotres-Alvarez D, Daviglus ML, Pirzada A, Mattei J, Perreira KM, Rotter JI, Taylor KD, Chen YDI, Wassertheil-Smoller S, Wang T, Rohan TE, Kaufman JD, Kaplan R, Qi Q. Genetic Subtypes of Prediabetes, Healthy Lifestyle, and Risk of Type 2 Diabetes. Diabetes 2024; 73:1178-1187. [PMID: 38602922 PMCID: PMC11189833 DOI: 10.2337/db23-0699] [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: 08/30/2023] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
Prediabetes is a heterogenous metabolic state with various risks for development of type 2 diabetes (T2D). In this study, we used genetic data on 7,227 US Hispanic/Latino participants without diabetes from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) and 400,149 non-Hispanic White participants without diabetes from the UK Biobank (UKBB) to calculate five partitioned polygenetic risk scores (pPRSs) representing various pathways related to T2D. Consensus clustering was performed in participants with prediabetes in HCHS/SOL (n = 3,677) and UKBB (n = 16,284) separately based on these pPRSs. Six clusters of individuals with prediabetes with distinctive patterns of pPRSs and corresponding metabolic traits were identified in the HCHS/SOL, five of which were confirmed in the UKBB. Although baseline glycemic traits were similar across clusters, individuals in cluster 5 and cluster 6 showed an elevated risk of T2D during follow-up compared with cluster 1 (risk ratios [RRs] 1.29 [95% CI 1.08, 1.53] and 1.34 [1.13, 1.60], respectively). Inverse associations between a healthy lifestyle score and risk of T2D were observed across different clusters, with a suggestively stronger association observed in cluster 5 compared with cluster 1. Among individuals with a healthy lifestyle, those in cluster 5 had a similar risk of T2D compared with those in cluster 1 (RR 1.03 [0.91, 1.18]). This study identified genetic subtypes of prediabetes that differed in risk of progression to T2D and in benefits from a healthy lifestyle. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yang Li
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Rhonda Arthur
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Daniela Sotres-Alvarez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Martha L. Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL
| | - Amber Pirzada
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL
| | - Josiemer Mattei
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Krista M. Perreira
- Department of Social Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | | | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Thomas E. Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Joel D. Kaufman
- Environmental and Occupational Health Sciences, Medicine, and Epidemiology, University of Washington, Seattle, WA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
31
|
EASL-EASD-EASO Clinical Practice Guidelines on the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Obes Facts 2024; 17:374-444. [PMID: 38852583 PMCID: PMC11299976 DOI: 10.1159/000539371] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
32
|
Kim SH. Reframing prediabetes: A call for better risk stratification and intervention. J Intern Med 2024; 295:735-747. [PMID: 38606904 DOI: 10.1111/joim.13786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Prediabetes is an intermediate state of glucose homeostasis whereby plasma glucose concentrations are above normal but below the threshold of diagnosis for diabetes. Over the last several decades, criteria for prediabetes have changed as the cut points for normal glucose concentration and diagnosis of diabetes have shifted. Global consensus does not exist for prediabetes criteria; as a result, the clinical course and risk for type 2 diabetes vary. At present, we can identify individuals with prediabetes based on three glycemic tests (hemoglobin A1c, fasting plasma glucose, and 2-h plasma glucose during an oral glucose tolerance test). The majority of individuals diagnosed with prediabetes meet only one of these criteria. Meeting one, two, or all glycemic criteria changes risk for type 2 diabetes, but this information is not widely known and does not currently guide intervention strategies for individuals with prediabetes. This review summarizes current epidemiology, prognosis, and intervention strategies for individuals diagnosed with prediabetes and suggests a call for more precise risk stratification of individuals with prediabetes as elevated (one prediabetes criterion), high risk (two prediabetes criteria), and very high risk (three prediabetes criteria). In addition, the roles of oral glucose tolerance testing and continuous glucose monitoring in the diagnostic criteria for prediabetes need reassessment. Finally, we must reframe our goals for prediabetes and prioritize intensive interventions for those at high and very high risk for type 2 diabetes.
Collapse
Affiliation(s)
- Sun H Kim
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
33
|
Nicolaisen SK, le Cessie S, Thomsen RW, Witte DR, Dekkers OM, Sørensen HT, Pedersen L. Longitudinal HbA1c patterns before the first treatment of diabetes in routine clinical practice: A latent class trajectory analysis. Diabetes Res Clin Pract 2024; 212:111722. [PMID: 38815656 DOI: 10.1016/j.diabres.2024.111722] [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: 11/10/2023] [Revised: 04/25/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
AIMS To examine the longitudinal heterogeneity of HbA1c preceding the initiation of diabetes treatment in clinical practice. METHODS In this population-based study, we used HbA1c from routine laboratory and healthcare databases. Latent class trajectory analysis was used to classify individuals according to their longitudinal HbA1c patterns before first glucose-lowering drug prescription irrespective of type of diabetes. RESULTS Among 21,556 individuals initiating diabetes treatment during 2017-2018, 20,733 (96 %) had HbA1c measured (median 4 measurements [IQR 2-7]) in the 5 years preceding treatment initiation. Four classes with distinct HbA1c trajectories were identified, with varying steepness of increase in HbA1c. The largest class (74 % of the individuals) had mean HbA1c above the 48 mmol/mol threshold 9 months before treatment initiation. Mean HbA1c was 52 mmol/mol (95 % CI 52-52) at treatment initiation. In the remaining three classes, mean HbA1c exceeded 48 mmol/mol almost 1.5 years before treatment initiation and reached 79 mmol/mol (95 % CI 78-80), 105 mmol/mol (95 % CI 104-106), and 137 mmol/mol (95 % CI 135-140) before treatment initiation. CONCLUSION We identified four distinct longitudinal HbA1c patterns before initiation of diabetes treatment in clinical practice. All had mean HbA1c levels exceeding the diagnostic threshold many months before treatment initiation, indicating therapeutic inertia.
Collapse
Affiliation(s)
- Sia Kromann Nicolaisen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark.
| | - Saskia le Cessie
- Department of Clinical Epidemiology & Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Reimar Wernich Thomsen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - Daniel R Witte
- Steno Diabetes Center Aarhus, Aarhus, Denmark; Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Olaf M Dekkers
- Department of Clinical Epidemiology & Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands
| | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - Lars Pedersen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| |
Collapse
|
34
|
Wang T, Shi Z, Ren H, Xu M, Lu J, Yang F, Ye C, Wu K, Chen M, Xu X, Liu D, Kong L, Zheng R, Zheng J, Li M, Xu Y, Zhao Z, Chen Y, Yang H, Wang J, Ning G, Li J, Zhong H, Bi Y, Wang W. Divergent age-associated and metabolism-associated gut microbiome signatures modulate cardiovascular disease risk. Nat Med 2024; 30:1722-1731. [PMID: 38844795 DOI: 10.1038/s41591-024-03038-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Insight into associations between the gut microbiome with metabolism and aging is crucial for tailoring interventions to promote healthy longevity. In a discovery cohort of 10,207 individuals aged 40-93 years, we used 21 metabolic parameters to classify individuals into five clusters, termed metabolic multimorbidity clusters (MCs), that represent different metabolic subphenotypes. Compared to the cluster classified as metabolically healthy (MC1), clusters classified as 'obesity-related mixed' (MC4) and 'hyperglycemia' (MC5) exhibited an increased 11.1-year cardiovascular disease (CVD) risk by 75% (multivariable-adjusted hazard ratio (HR): 1.75, 95% confidence interval (CI): 1.43-2.14) and by 117% (2.17, 1.72-2.74), respectively. These associations were replicated in a second cohort of 9,061 individuals with a 10.0-year follow-up. Based on analysis of 4,491 shotgun fecal metagenomes from the discovery cohort, we found that gut microbial composition was associated with both MCs and age. Next, using 55 age-specific microbial species to capture biological age, we developed a gut microbial age (MA) metric, which was validated in four external cohorts comprising 4,425 metagenomic samples. Among individuals aged 60 years or older, the increased CVD risk associated with MC4 or MC5, as compared to MC1, MC2 or MC3, was exacerbated in individuals with high MA but diminished in individuals with low MA, independent of age, sex and other lifestyle and dietary factors. This pattern, in which younger MA appears to counteract the CVD risk attributable to metabolic dysfunction, implies a modulating role of MA in cardiovascular health for metabolically unhealthy older people.
Collapse
Affiliation(s)
- Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhun Shi
- BGI Research, Shenzhen, China
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
| | - Huahui Ren
- BGI Research, Shenzhen, China
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Chaojie Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Wu
- BGI Research, Shenzhen, China
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, China
| | - Mingling Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xun Xu
- BGI Research, Shenzhen, China
| | - Dong Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijie Kong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Huanzi Zhong
- BGI Research, Shenzhen, China.
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
35
|
Fermín-Martínez CA, Bello-Chavolla OY, Paz-Cabrera CD, Ramírez-García D, Perezalonso-Espinosa J, Fernández-Chirino L, Vargas-Vázquez A, Díaz-Sánchez JP, Méndez-Labra PN, Núñez-Luna A, Basile-Alvarez MR, Sánchez-Castro P, Bragg F, Friedrichs LG, Aguilar-Ramírez D, Emberson JR, Berumen-Campos J, Kuri-Morales P, Tapia-Conyer R, Alegre-Díaz J, Seiglie JA, Antonio-Villa NE. Prediabetes as a risk factor for all-cause and cause-specific mortality: a prospective analysis of 115,919 adults without diabetes in Mexico City. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.15.24305840. [PMID: 38699295 PMCID: PMC11065040 DOI: 10.1101/2024.04.15.24305840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
BACKGROUND Prediabetes has been associated with increased all-cause and cardiovascular mortality. However, no large-scale studies have been conducted in Mexico or Latin America examining these associations. METHODS We analyzed data from 115,919 adults without diabetes (diagnosed or undiagnosed) aged 35-84 years who participated in the Mexico City Prospective Study between 1998 and 2004. Participants were followed until January 1st, 2021 for cause-specific mortality. We defined prediabetes according to the American Diabetes Association (ADA, HbA1c 5.7% to 6.4%) and the International Expert Committee (IEC, HbA1c 6.0-6.4%) definitions. Cox regression adjusted for confounders was used to estimate all-cause and cause-specific mortality rate ratios (RR) at ages 35-74 years associated with prediabetes. FINDINGS During 2,085,392 person-years of follow-up (median in survivors 19 years), there were 6,810 deaths at ages 35-74, including 1,742 from cardiovascular disease, 892 from renal disease and 108 from acute diabetic crises. Of 110,405 participants aged 35-74 years at recruitment, 28,852 (26%) had ADA-defined prediabetes and 7,203 (7%) had IEC-defined prediabetes. Compared with those without prediabetes, individuals with prediabetes had higher risk of all-cause mortality at ages 35-74 years (RR 1.13, 95% CI 1.07-1.19 for ADA-defined prediabetes and RR 1.28, 1.18-1.39 for IEC-defined prediabetes), as well as increased risk of cardiovascular mortality (RR 1.22 [1.10-1.35] and 1.42 [1.22-1.65], respectively), renal mortality (RR 1.35 [1.08-1.68] and 1.69 [1.24-2.31], respectively), and death from an acute diabetic crisis (RR 2.63 [1.76-3.94] and 3.43 [2.09-5.62], respectively). RRs were larger at younger than at older ages, and similar for men compared to women. The absolute excess risk associated with ADA and IEC-defined prediabetes at ages 35-74 accounted for6% and 3% of cardiovascular deaths respectively, 10% and 5% of renal deaths respectively, and 31% and 14% of acute diabetic deaths respectively. INTERPRETATION Prediabetes is a significant risk factor for all-cause, cardiovascular, renal, and acute diabetic deaths in Mexican adults. Identification and timely management of individuals with prediabetes for targeted risk reduction could contribute to reducing premature mortality from cardiometabolic causes in this population. FUNDING Wellcome Trust, the Mexican Health Ministry, the National Council of Science and Technology for Mexico, Cancer Research UK, British Heart Foundation, UK Medical Research Council. Instituto Nacional de Geriatría (Mexico City).
Collapse
Affiliation(s)
- Carlos A. Fermín-Martínez
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - César Daniel Paz-Cabrera
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- Especialidad en Medicina Preventiva, Instituto Nacional de Salud Pública, Mexico City, Mexico
| | - Daniel Ramírez-García
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jerónimo Perezalonso-Espinosa
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luisa Fernández-Chirino
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Arsenio Vargas-Vázquez
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juan Pablo Díaz-Sánchez
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Padme Nailea Méndez-Labra
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandra Núñez-Luna
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Martín Roberto Basile-Alvarez
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paulina Sánchez-Castro
- Research Division, Instituto Nacional de Geriatría, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Fiona Bragg
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Louisa Gnatiuc Friedrichs
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Diego Aguilar-Ramírez
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jonathan R. Emberson
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jaime Berumen-Campos
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Pablo Kuri-Morales
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Roberto Tapia-Conyer
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jesus Alegre-Díaz
- Experimental Research Unit, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Jacqueline A. Seiglie
- Diabetes Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
36
|
Parveen A, Batool A, Wajid A, Mukhtar M, Khan KU, Zahid A, Jabeen A, Sahibzada KI. Delving the vitamin D receptor variation and expression profiles in the context of type 2 diabetes among families. Mol Biol Rep 2024; 51:514. [PMID: 38622480 DOI: 10.1007/s11033-024-09387-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/26/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Vitamin D is essential for insulin secretion and sensitivity. Consequently, its inadequacy is linked to higher insulin resistance and Type 2 Diabetes (T2D). The Vitamin D receptor (VDR) gene is one potential candidate for T2D, and multiple polymorphisms in VDR have been examined in various populations, but no conclusive answers have been provided. OBJECTIVES This study was designed to evaluate the susceptibility of VDR gene polymorphism and its expression in diabetic families in Pakistan. METHODOLOGY In this family-based study, twenty diabetic families with a positive family history of T2D and at least three T2D patients were recruited from outpatient clinics and public hospitals. The current study comprised 143 individuals with 55 affected and 88 unaffected individuals. Blood samples of the selected families were collected. DNA was extracted from the collected samples and the PCR-RFLP method was followed to identify the genotyping and RT-qPCR for expression. Phenotypic and genotypic pedigrees of the families were developed by the progeny online tool. The association values of SNPs were determined by TDT and DFAM analysis implemented on Plink software. RESULTS The results explained a significant familial aggregation among phenotypic characters including Age, Gender, BMI (body mass index), age of disease diagnosis, disease duration, and blood pressure in the probands, affected FDRs (First Degree Relatives) and affected SDRs (Second Degree Relatives). A significant association of rs731236 C/T (OR = 1.522), rs2228570 C/T (OR = 1.327) with p < 0.05. Whereas, for rs1544410 G/A (OR = 0.9706) and rs7975232 T/G (OR = 0.7368) no considerable association evidence was seen (p > 0.05) in families. The mRNA expression of VDR increased threefold (p = 0.0204) in patients compared to controls. Variation-based expression analysis exhibited that the rs2228570 genotype influences the expression. CONCLUSION A linkage was found among the FDRs with probands. Variation in the gene VDR at loci rs731236 and rs2228570 was associated with familial T2D. However further research is required to explore more genetic factors that could influence T2D risks in families.
Collapse
Affiliation(s)
- Asia Parveen
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Andleeb Batool
- Department of Zoology, Government College University, Lahore, Pakistan.
| | - Abdul Wajid
- Balochistan University of Information Technology Engineering and Management Sciences, Quetta, Pakistan
| | - Maryam Mukhtar
- Department of Zoology, Government College University, Lahore, Pakistan
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Khajid Ullah Khan
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Aqsa Zahid
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Anjum Jabeen
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Kashif Iqbal Sahibzada
- Department of Health Professional Technologies, Faculty of Allied Health Sciences, University of Lahore, Lahore, Pakistan
- School of Bioengineering, Henan University of Technology, Zhengzhou, China
| |
Collapse
|
37
|
Patel CJ, Ioannidis JP, Gregg EW, Vasan RS, Manrai AK. Heterogeneity in elevated glucose and A1C as predictors of the prediabetes to diabetes transition: Framingham Heart Study, Multi-Ethnic Study on Atherosclerosis, Jackson Heart Study, and Atherosclerosis Risk In Communities. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.16.24304398. [PMID: 38562763 PMCID: PMC10984063 DOI: 10.1101/2024.03.16.24304398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Introduction There are a number of glycemic definitions for prediabetes; however, the heterogeneity in diabetes transition rates from prediabetes across different glycemic definitions in major US cohorts has been unexplored. We estimate the variability in risk and relative risk of adiposity based on diagnostic criteria like fasting glucose and hemoglobin A1C% (HA1C%). Research Design and Methods We estimated transition rate from prediabetes, as defined by fasting glucose between 100-125 and/or 110-125 mg/dL, and HA1C% between 5.7-6.5% in participant data from the Framingham Heart Study, Multi-Ethnic Study on Atherosclerosis, Atherosclerosis Risk in Communities, and the Jackson Heart Study. We estimated the heterogeneity and prediction interval across cohorts, stratifying by age, sex, and body mass index. For individuals who were prediabetic, we estimated the relative risk for obesity, blood pressure, education, age, and sex for diabetes. Results There is substantial heterogeneity in diabetes transition rates across cohorts and prediabetes definitions with large prediction intervals. We observed the highest range of rates in individuals with fasting glucose of 110-125 mg/dL ranging from 2-18 per 100 person-years. Across different cohorts, the association obesity or hypertension in the progression to diabetes was consistent, yet it varied in magnitude. We provide a database of transition rates across subgroups and cohorts for comparison in future studies. Conclusion The absolute transition rate from prediabetes to diabetes significantly depends on cohort and prediabetes definitions.
Collapse
Affiliation(s)
- Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA. 02215
| | - John Pa Ioannidis
- Department of Prevention Research, Stanford University School of Medicine, Stanford, CA. 94305
| | - Edward W Gregg
- School of Population Health, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Arjun K Manrai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA. 02215
| |
Collapse
|
38
|
Washirasaksiri C, Pakornnipat W, Ariyakunaphan P, Kositamongkol C, Polmanee C, Preechasuk L, Jaiborisuttigull N, Sitasuwan T, Tinmanee R, Pramyothin P, Srivanichakorn W. Effectiveness of a cognitive behavioral therapy-integrated, hospital-based program for prediabetes: a matched cohort study. Sci Rep 2024; 14:8010. [PMID: 38580745 PMCID: PMC10997588 DOI: 10.1038/s41598-024-58739-8] [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: 12/11/2023] [Accepted: 04/02/2024] [Indexed: 04/07/2024] Open
Abstract
Intensive lifestyle interventions are effective in preventing T2DM, but evidence is lacking for high cardiometabolic individuals in hospital settings. We evaluated a hospital-based, diabetes prevention program integrating cognitive behavioral therapy (CBT) for individuals with prediabetes. This matched cohort assessed individuals with prediabetes receiving the prevention program, which were matched 1:1 with those receiving standard care. The year-long program included five in-person sessions and several online sessions covering prediabetes self-management, dietary and behavioral interventions. Kaplan-Meier and Cox regression models estimated the 60-month T2DM incidence rate. Of 192 patients, 190 joined the prevention program, while 190 out of 10,260 individuals were in the standard-care group. Both groups had similar baseline characteristics (mean age 58.9 ± 10.2 years, FPG 102.3 ± 8.2 mg/dL, HbA1c 5.9 ± 0.3%, BMI 26.2 kg/m2, metabolic syndrome 75%, and ASCVD 6.3%). After 12 months, the intervention group only showed significant decreases in FPG, HbA1c, and triglyceride levels and weight. At 60 months, the T2DM incidence rate was 1.7 (95% CI 0.9-2.8) in the intervention group and 3.5 (2.4-4.9) in the standard-care group. After adjusting for variables, the intervention group had a 0.46 times lower risk of developing diabetes. Therefore, healthcare providers should actively promote CBT-integrated, hospital-based diabetes prevention programs to halve diabetes progression.
Collapse
Affiliation(s)
- Chaiwat Washirasaksiri
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wang Lang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Withada Pakornnipat
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pinyapat Ariyakunaphan
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wang Lang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Chayanis Kositamongkol
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wang Lang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Chaiyaporn Polmanee
- Siriraj Diabetes Center of Excellence, Mahidol University, Bangkok, Thailand
| | - Lukana Preechasuk
- Siriraj Diabetes Center of Excellence, Mahidol University, Bangkok, Thailand
| | - Naris Jaiborisuttigull
- Preventive and Health Promotion Nursing Unit, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tullaya Sitasuwan
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wang Lang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Rungsima Tinmanee
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wang Lang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Pornpoj Pramyothin
- Division of Nutrition, Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Weerachai Srivanichakorn
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wang Lang Road, Bangkok Noi, Bangkok, 10700, Thailand.
- Siriraj Diabetes Center of Excellence, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
39
|
Muse ED, Topol EJ. Transforming the cardiometabolic disease landscape: Multimodal AI-powered approaches in prevention and management. Cell Metab 2024; 36:670-683. [PMID: 38428435 PMCID: PMC10990799 DOI: 10.1016/j.cmet.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
The rise of artificial intelligence (AI) has revolutionized various scientific fields, particularly in medicine, where it has enabled the modeling of complex relationships from massive datasets. Initially, AI algorithms focused on improved interpretation of diagnostic studies such as chest X-rays and electrocardiograms in addition to predicting patient outcomes and future disease onset. However, AI has evolved with the introduction of transformer models, allowing analysis of the diverse, multimodal data sources existing in medicine today. Multimodal AI holds great promise in more accurate disease risk assessment and stratification as well as optimizing the key driving factors in cardiometabolic disease: blood pressure, sleep, stress, glucose control, weight, nutrition, and physical activity. In this article we outline the current state of medical AI in cardiometabolic disease, highlighting the potential of multimodal AI to augment personalized prevention and treatment strategies in cardiometabolic disease.
Collapse
Affiliation(s)
- Evan D Muse
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA 92037, USA; Division of Cardiovascular Diseases, Scripps Clinic, La Jolla, CA 92037, USA
| | - Eric J Topol
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA 92037, USA; Division of Cardiovascular Diseases, Scripps Clinic, La Jolla, CA 92037, USA.
| |
Collapse
|
40
|
Heilmann G, Trenkamp S, Möser C, Bombrich M, Schön M, Yurchenko I, Strassburger K, Rodríguez MM, Zaharia OP, Burkart V, Wagner R, Roden M. Precise glucose measurement in sodium fluoride-citrate plasma affects estimates of prevalence in diabetes and prediabetes. Clin Chem Lab Med 2024; 62:762-769. [PMID: 37870928 DOI: 10.1515/cclm-2023-0770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023]
Abstract
OBJECTIVES Estimates of glucose concentrations vary among types of blood samples, which impact on the assessment of diabetes prevalence. Guidelines recommend a conversion factor to calculate plasma glucose from measurements of glucose in whole blood. The American Diabetes Association recommends the use of blood drawing tubes containing sodium fluoride (NaF) and citrate, which have not yet been evaluated regarding possible differences in glucose concentration and conversion factors. Thus, we compared glucose measurements in NaF-citrate plasma and venous whole blood and estimated the impact of differences on diabetes and prediabetes prevalence. METHODS Glucose differences were calculated by Bland-Altman analysis with pairwise comparison of glucose measurements from whole blood and NaF-citrate plasma (n=578) in clinical studies of the German Diabetes Center. Subsequently, we computed the impact of the glucose difference on diabetes and prediabetes prevalence in the population-based National Health and Nutrition Examination Survey (NHANES). RESULTS Even upon conversion of whole blood to plasma glucose concentrations using the recommended conversion factor, mean glucose concentration difference remained 4.72 % higher in NaF-citrate plasma. Applying the higher glucose estimates, increases the population-based diabetes and prediabetes prevalence by 13.67 and 33.97 % or more than 7.2 and 13 million people in NHANES, respectively. Additional economic burden could be about 20 $ billion per year due to undiagnosed diabetes. CONCLUSIONS The recommended conversion factor is not valid for NaF-citrate plasma. Systematic bias of glucose measurements due to sampling type leads to clinically relevant higher estimates of diabetes and prediabetes prevalence.
Collapse
Affiliation(s)
- Geronimo Heilmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Sandra Trenkamp
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Clara Möser
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Maria Bombrich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Iryna Yurchenko
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
| | - Marcos Matabuena Rodríguez
- Centro Singular de Investigación en Tecnoloxías Intelixentes, Universidade de Santiago de Compostela, Santiago, Spain
| | - Oana-Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
41
|
Li S, Cui M, Liu Y, Liu X, Luo L, Zhao W, Gu X, Li L, Liu C, Bai L, Li D, Liu B, Che D, Li X, Wang Y, Gao Z. Metabolic Profiles of Type 2 Diabetes and Their Association With Renal Complications. J Clin Endocrinol Metab 2024; 109:1051-1059. [PMID: 37933705 DOI: 10.1210/clinem/dgad643] [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: 07/19/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/08/2023]
Abstract
CONTEXT The components of metabolic syndrome (MetS) are interrelated and associated with renal complications in patients with type 2 diabetes (T2D). OBJECTIVE We aimed to reveal prevalent metabolic profiles in patients with T2D and identify which metabolic profiles were risk markers for renal progression. METHODS A total of 3556 participants with T2D from a hospital (derivation cohort) and 931 participants with T2D from a community survey (external validation cohort) were included. The primary outcome was the onset of diabetic kidney disease (DKD), and secondary outcomes included estimated glomerular filtration rate (eGFR) decline, macroalbuminuria, and end-stage renal disease (ESRD). In the derivation cohort, clusters were identified using the 5 components of MetS, and their relationships with the outcomes were assessed. To validate the findings, participants in the validation cohort were assigned to clusters. Multivariate odds ratios (ORs) of the primary outcome were evaluated in both cohorts, adjusted for multiple covariates at baseline. RESULTS In the derivation cohort, 6 clusters were identified as metabolic profiles. Compared with cluster 1, cluster 3 (severe hyperglycemia) had increased risks of DKD (hazard ratio [HR] [95% CI]: 1.72 [1.39-2.12]), macroalbuminuria (2.74 [1.84-4.08]), ESRD (4.31 [1.16-15.99]), and eGFR decline [P < .001]; cluster 4 (moderate dyslipidemia) had increased risks of DKD (1.97 [1.53-2.54]) and macroalbuminuria (2.62 [1.61-4.25]). In the validation cohort, clusters 3 and 4 were replicated to have significantly increased risks of DKD (adjusted ORs: 1.24 [1.07-1.44] and 1.39 [1.03-1.87]). CONCLUSION We identified 6 prevalent metabolic profiles in patients with T2D. Severe hyperglycemia and moderate dyslipidemia were validated as significant risk markers for DKD.
Collapse
Affiliation(s)
- Shen Li
- Department of Central Laboratory, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Mengxuan Cui
- Yidu Cloud Technology Inc, Beijing 100101, China
| | - Yingshu Liu
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Xuhan Liu
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Lan Luo
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Wei Zhao
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Xiaolan Gu
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Linfeng Li
- Yidu Cloud Technology Inc, Beijing 100101, China
| | - Chao Liu
- Yidu Cloud Technology Inc, Beijing 100101, China
| | - Lan Bai
- Yidu Cloud Technology Inc, Beijing 100101, China
| | - Di Li
- Department of Neurointervention, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Bo Liu
- School of Biomedical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Defei Che
- Department of Medical Equipment, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Xinyu Li
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| | - Yao Wang
- Yidu Cloud Technology Inc, Beijing 100101, China
| | - Zhengnan Gao
- Department of Endocrinology, Central Hospital of Dalian University of Technology, Dalian 116000, China
| |
Collapse
|
42
|
Kim NY, Lee H, Kim S, Kim YJ, Lee H, Lee J, Kwak SH, Lee S. The clinical relevance of a polygenic risk score for type 2 diabetes mellitus in the Korean population. Sci Rep 2024; 14:5749. [PMID: 38459065 PMCID: PMC10923897 DOI: 10.1038/s41598-024-55313-0] [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: 05/30/2023] [Accepted: 02/22/2024] [Indexed: 03/10/2024] Open
Abstract
The clinical utility of a type 2 diabetes mellitus (T2DM) polygenic risk score (PRS) in the East Asian population remains underexplored. We aimed to examine the potential prognostic value of a T2DM PRS and assess its viability as a clinical instrument. We first established a T2DM PRS for 5490 Korean individuals using East Asian Biobank data (269,487 samples). Subsequently, we assessed the predictive capability of this T2DM PRS in a prospective longitudinal study with baseline data and data from seven additional follow-ups. Our analysis showed that the T2DM PRS could predict the transition of glucose tolerance stages from normal glucose tolerance to prediabetes and from prediabetes to T2DM. Moreover, T2DM patients in the top-decile PRS group were more likely to be treated with insulin (hazard ratio = 1.69, p value = 2.31E-02) than were those in the remaining PRS groups. T2DM PRS values were significantly high in the severe diabetes subgroup, characterized by insulin resistance and β -cell dysfunction (p value = 0.0012). The prediction models with the T2DM PRS had significantly greater Harrel's C-indices than did corresponding models without it. By utilizing prospective longitudinal study data and extensive clinical risk factor information, our analysis provides valuable insights into the multifaceted clinical utility of the T2DM PRS.
Collapse
Affiliation(s)
- Na Yeon Kim
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Haekyung Lee
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, South Korea
| | - Sehee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, Seoul, South Korea
| | - Ye-Jee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, Seoul, South Korea
| | - Hyunsuk Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Junhyeong Lee
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Seunggeun Lee
- Graduate School of Data Science, Seoul National University, Seoul, South Korea.
| |
Collapse
|
43
|
Pleus S, Tytko A, Landgraf R, Heinemann L, Werner C, Müller-Wieland D, Ziegler AG, Müller UA, Freckmann G, Kleinwechter H, Schleicher E, Nauck M, Petersmann A. Definition, Classification, Diagnosis and Differential Diagnosis of Diabetes Mellitus: Update 2023. Exp Clin Endocrinol Diabetes 2024; 132:112-124. [PMID: 38378016 DOI: 10.1055/a-2166-6643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Affiliation(s)
- Stefan Pleus
- Institut für Diabetes-Technologie Forschungs- und Entwicklungsgesellschaft mbH an der Universität Ulm, Ulm, Germany
| | | | | | - Lutz Heinemann
- Science-Consulting in Diabetes GmbH, Düsseldorf, Germany
| | - Christoph Werner
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | | | | | - Ulrich A Müller
- Practice for Endocrinology and Diabetology, Dr. Kielstein Ambulante Medizinische Versorgung GmbH, Jena, Germany
| | - Guido Freckmann
- Institut für Diabetes-Technologie Forschungs- und Entwicklungsgesellschaft mbH an der Universität Ulm, Ulm, Germany
| | | | - Erwin Schleicher
- Institute of Clinical Chemistry and Pathobiochemistry - Central Laboratory, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD) Munich-Neuherberg, Munich-Neuherberg, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Oldenburg, Oldenburg, Germany
| |
Collapse
|
44
|
Lizarzaburu-Robles JC, Herman WH, Garro-Mendiola A, Galdón Sanz-Pastor A, Lorenzo O. Prediabetes and Cardiometabolic Risk: The Need for Improved Diagnostic Strategies and Treatment to Prevent Diabetes and Cardiovascular Disease. Biomedicines 2024; 12:363. [PMID: 38397965 PMCID: PMC10887025 DOI: 10.3390/biomedicines12020363] [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: 12/14/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
The progression from prediabetes to type-2 diabetes depends on multiple pathophysiological, clinical, and epidemiological factors that generally overlap. Both insulin resistance and decreased insulin secretion are considered to be the main causes. The diagnosis and approach to the prediabetic patient are heterogeneous. There is no agreement on the diagnostic criteria to identify prediabetic subjects or the approach to those with insufficient responses to treatment, with respect to regression to normal glycemic values or the prevention of complications. The stratification of prediabetic patients, considering the indicators of impaired fasting glucose, impaired glucose tolerance, or HbA1c, can help to identify the sub-phenotypes of subjects at risk for T2DM. However, considering other associated risk factors, such as impaired lipid profiles, or risk scores, such as the Finnish Diabetes Risk Score, may improve classification. Nevertheless, we still do not have enough information regarding cardiovascular risk reduction. The sub-phenotyping of subjects with prediabetes may provide an opportunity to improve the screening and management of cardiometabolic risk in subjects with prediabetes.
Collapse
Affiliation(s)
- Juan Carlos Lizarzaburu-Robles
- Endocrinology Unit, Hospital Central de la Fuerza Aérea del Perú, 15046 Lima, Peru;
- Doctorate Program, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - William H. Herman
- Department of Internal Medicine and Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | | | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma, 28049 Madrid, Spain;
- Biomedical Research Network on Diabetes and Associated Metabolic Disorders (CIBERDEM), Carlos III National Health Institute, 28029 Madrid, Spain
| |
Collapse
|
45
|
Ungvari Z, Tabák AG, Adany R, Purebl G, Kaposvári C, Fazekas-Pongor V, Csípő T, Szarvas Z, Horváth K, Mukli P, Balog P, Bodizs R, Ujma P, Stauder A, Belsky DW, Kovács I, Yabluchanskiy A, Maier AB, Moizs M, Östlin P, Yon Y, Varga P, Vokó Z, Papp M, Takács I, Vásárhelyi B, Torzsa P, Ferdinandy P, Csiszar A, Benyó Z, Szabó AJ, Dörnyei G, Kivimäki M, Kellermayer M, Merkely B. The Semmelweis Study: a longitudinal occupational cohort study within the framework of the Semmelweis Caring University Model Program for supporting healthy aging. GeroScience 2024; 46:191-218. [PMID: 38060158 PMCID: PMC10828351 DOI: 10.1007/s11357-023-01018-7] [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: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
The Semmelweis Study is a prospective occupational cohort study that seeks to enroll all employees of Semmelweis University (Budapest, Hungary) aged 25 years and older, with a population of 8866 people, 70.5% of whom are women. The study builds on the successful experiences of the Whitehall II study and aims to investigate the complex relationships between lifestyle, environmental, and occupational risk factors, and the development and progression of chronic age-associated diseases. An important goal of the Semmelweis Study is to identify groups of people who are aging unsuccessfully and therefore have an increased risk of developing age-associated diseases. To achieve this, the study takes a multidisciplinary approach, collecting economic, social, psychological, cognitive, health, and biological data. The Semmelweis Study comprises a baseline data collection with open healthcare data linkage, followed by repeated data collection waves every 5 years. Data are collected through computer-assisted self-completed questionnaires, followed by a physical health examination, physiological measurements, and the assessment of biomarkers. This article provides a comprehensive overview of the Semmelweis Study, including its origin, context, objectives, design, relevance, and expected contributions.
Collapse
Affiliation(s)
- Zoltan Ungvari
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Adam G Tabák
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Roza Adany
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-UD Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Purebl
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Csilla Kaposvári
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Csípő
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Szarvas
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Krisztián Horváth
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Mukli
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Piroska Balog
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Robert Bodizs
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Ujma
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Adrienne Stauder
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Daniel W Belsky
- Robert N. Butler Columbia Aging Center, Columbia University, New York, NY, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Illés Kovács
- Department of Ophthalmology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA
- Department of Clinical Ophthalmology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System, Singapore, Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Vrije Universiteit, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Mariann Moizs
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Ministry of Interior of Hungary, Budapest, Hungary
| | | | - Yongjie Yon
- WHO Regional Office for Europe, Copenhagen, Denmark
| | - Péter Varga
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Clinical Center, Semmelweis University, Budapest, Hungary
| | - Zoltán Vokó
- Center for Health Technology Assessment, Semmelweis University, Budapest, Hungary
| | - Magor Papp
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - István Takács
- UCL Brain Sciences, University College London, London, UK
| | - Barna Vásárhelyi
- Department of Laboratory Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Torzsa
- Department of Family Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltán Benyó
- Department of Translational Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, Budapest, Hungary
| | - Attila J Szabó
- First Department of Pediatrics, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Pediatrics and Nephrology Research Group, Semmelweis University, Budapest, Hungary
| | - Gabriella Dörnyei
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Mika Kivimäki
- UCL Brain Sciences, University College London, London, UK
| | - Miklos Kellermayer
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
46
|
Schön M, Prystupa K, Mori T, Zaharia OP, Bódis K, Bombrich M, Möser C, Yurchenko I, Kupriyanova Y, Strassburger K, Bobrov P, Nair ATN, Bönhof GJ, Strom A, Delgado GE, Kaya S, Guthoff R, Stefan N, Birkenfeld AL, Hauner H, Seissler J, Pfeiffer A, Blüher M, Bornstein S, Szendroedi J, Meyhöfer S, Trenkamp S, Burkart V, Schrauwen-Hinderling VB, Kleber ME, Niessner A, Herder C, Kuss O, März W, Pearson ER, Roden M, Wagner R. Analysis of type 2 diabetes heterogeneity with a tree-like representation: insights from the prospective German Diabetes Study and the LURIC cohort. Lancet Diabetes Endocrinol 2024; 12:119-131. [PMID: 38142707 DOI: 10.1016/s2213-8587(23)00329-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Heterogeneity in type 2 diabetes can be represented by a tree-like graph structure by use of reversed graph-embedded dimensionality reduction. We aimed to examine whether this approach can be used to stratify key pathophysiological components and diabetes-related complications during longitudinal follow-up of individuals with recent-onset type 2 diabetes. METHODS For this cohort analysis, 927 participants aged 18-69 years from the German Diabetes Study (GDS) with recent-onset type 2 diabetes were mapped onto a previously developed two-dimensional tree based on nine simple clinical and laboratory variables, residualised for age and sex. Insulin sensitivity was assessed by a hyperinsulinaemic-euglycaemic clamp, insulin secretion was assessed by intravenous glucose tolerance test, hepatic lipid content was assessed by 1 H magnetic resonance spectroscopy, serum interleukin (IL)-6 and IL-18 were assessed by ELISA, and peripheral and autonomic neuropathy were assessed by functional and clinical measures. Participants were followed up for up to 16 years. We also investigated heart failure and all-cause mortality in 794 individuals with type 2 diabetes undergoing invasive coronary diagnostics from the Ludwigshafen Risk and Cardiovascular Health (LURIC) cohort. FINDINGS There were gradients of clamp-measured insulin sensitivity (both dimensions: p<0·0001) and insulin secretion (pdim1<0·0001, pdim2=0·00097) across the tree. Individuals in the region with the lowest insulin sensitivity had the highest hepatic lipid content (n=205, pdim1<0·0001, pdim2=0·037), pro-inflammatory biomarkers (IL-6: n=348, pdim1<0·0001, pdim2=0·013; IL-18: n=350, pdim1<0·0001, pdim2=0·38), and elevated cardiovascular risk (nevents=143, pdim1=0·14, pdim2<0·00081), whereas individuals positioned in the branch with the lowest insulin secretion were more prone to require insulin therapy (nevents=85, pdim1=0·032, pdim2=0·12) and had the highest risk of diabetic sensorimotor polyneuropathy (nevents=184, pdim1=0·012, pdim2=0·044) and cardiac autonomic neuropathy (nevents=118, pdim1=0·0094, pdim2=0·06). In the LURIC cohort, all-cause mortality was highest in the tree branch showing insulin resistance (nevents=488, pdim1=0·12, pdim2=0·0032). Significant gradients differentiated individuals having heart failure with preserved ejection fraction from those who had heart failure with reduced ejection fraction. INTERPRETATION These data define the pathophysiological underpinnings of the tree structure, which has the potential to stratify diabetes-related complications on the basis of routinely available variables and thereby expand the toolbox of precision diabetes diagnosis. FUNDING German Diabetes Center, German Federal Ministry of Health, Ministry of Culture and Science of the state of North Rhine-Westphalia, German Federal Ministry of Education and Research, German Diabetes Association, German Center for Diabetes Research, European Community, German Research Foundation, and Schmutzler Stiftung.
Collapse
Affiliation(s)
- Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Tim Mori
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Oana P Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maria Bombrich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Clara Möser
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Iryna Yurchenko
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Anand T N Nair
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Graciela E Delgado
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Preventive Medicine and Digital Health Baden-Württemberg, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sema Kaya
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Rainer Guthoff
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, München, Germany
| | - Jochen Seissler
- Diabetes Research Group, Medical Department 4, Ludwig-Maximilians University Munich, München, Germany
| | - Andreas Pfeiffer
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Matthias Blüher
- Department of Medicine, Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Stefan Bornstein
- Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Julia Szendroedi
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Svenja Meyhöfer
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany; Department of Internal Medicine 1, Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany
| | - Sandra Trenkamp
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Vera B Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Marcus E Kleber
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Alexander Niessner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Austria
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver Kuss
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Winfried März
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
| | - Ewan R Pearson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
47
|
Szczerbinski L, Florez JC. Precision medicine in diabetes - current trends and future directions. Is the future now? COMPREHENSIVE PRECISION MEDICINE 2024:458-483. [DOI: 10.1016/b978-0-12-824010-6.00021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
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
|
49
|
Muniyappa R, Narayanappa SBK. Disentangling Dual Threats: Premature Coronary Artery Disease and Early-Onset Type 2 Diabetes Mellitus in South Asians. J Endocr Soc 2023; 8:bvad167. [PMID: 38178904 PMCID: PMC10765382 DOI: 10.1210/jendso/bvad167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Indexed: 01/06/2024] Open
Abstract
South Asian individuals (SAs) face heightened risks of premature coronary artery disease (CAD) and early-onset type 2 diabetes mellitus (T2DM), with grave health, societal, and economic implications due to the region's dense population. Both conditions, influenced by cardiometabolic risk factors such as insulin resistance, hypertension, and central adiposity, manifest earlier and with unique thresholds in SAs. Epidemiological, demographic, nutritional, environmental, sociocultural, and economic transitions in SA have exacerbated the twin epidemic. The coupling of premature CAD and T2DM arises from increased obesity due to limited adipose storage, early-life undernutrition, distinct fat thresholds, reduced muscle mass, and a predisposition for hepatic fat accumulation from certain dietary choices cumulatively precipitating a decline in insulin sensitivity. As T2DM ensues, the β-cell adaptive responses are suboptimal, precipitating a transition from compensatory hyperinsulinemia to β-cell decompensation, underscoring a reduced functional β-cell reserve in SAs. This review delves into the interplay of these mechanisms and highlights a prediabetes endotype tied to elevated vascular risk. Deciphering these mechanistic interconnections promises to refine stratification paradigms, surpassing extant risk-prediction strategies.
Collapse
Affiliation(s)
- Ranganath Muniyappa
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Satish Babu K Narayanappa
- Department of Medicine, Sri Madhusudan Sai Institute of Medical Sciences and Research, Muddenahalli, Karnataka 562101, India
| |
Collapse
|
50
|
Hillesheim E, Brennan L. Distinct patterns of personalised dietary advice delivered by a metabotype framework similarly improve dietary quality and metabolic health parameters: secondary analysis of a randomised controlled trial. Front Nutr 2023; 10:1282741. [PMID: 38035361 PMCID: PMC10684740 DOI: 10.3389/fnut.2023.1282741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Background In a 12-week randomised controlled trial, personalised nutrition delivered using a metabotype framework improved dietary intake, metabolic health parameters and the metabolomic profile compared to population-level dietary advice. The objective of the present work was to investigate the patterns of dietary advice delivered during the intervention and the alterations in dietary intake and metabolic and metabolomic profiles to obtain further insights into the effectiveness of the metabotype framework. Methods Forty-nine individuals were randomised into the intervention group and subsequently classified into metabotypes using four biomarkers (triacylglycerol, HDL-C, total cholesterol, glucose). These individuals received personalised dietary advice from decision tree algorithms containing metabotypes and individual characteristics. In a secondary analysis of the data, patterns of dietary advice were identified by clustering individuals according to the dietary messages received and clusters were compared for changes in dietary intake and metabolic health parameters. Correlations between changes in blood clinical chemistry and changes in metabolite levels were investigated. Results Two clusters of individuals with distinct patterns of dietary advice were identified. Cluster 1 had the highest percentage of messages delivered to increase the intake of beans and pulses and milk and dairy products. Cluster 2 had the highest percentage of messages delivered to limit the intake of foods high in added sugar, high-fat foods and alcohol. Following the intervention, both patterns improved dietary quality assessed by the Alternate Mediterranean Diet Score and the Alternative Healthy Eating Index, nutrient intakes, blood pressure, triacylglycerol and LDL-C (p ≤ 0.05). Several correlations were identified between changes in total cholesterol, LDL-C, triacylglycerol, insulin and HOMA-IR and changes in metabolites levels, including mostly lipids (sphingomyelins, lysophosphatidylcholines, glycerophosphocholines and fatty acid carnitines). Conclusion The findings indicate that the metabotype framework effectively personalises and delivers dietary advice to improve dietary quality and metabolic health. Clinical trial registration isrctn.com, identifier ISRCTN15305840.
Collapse
Affiliation(s)
- Elaine Hillesheim
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Lorraine Brennan
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|