1
|
Sasaki N, Ueno Y, Ozono R, Nakano Y, Higashi Y. Insulin resistance in adipose tissue and fatty liver, but not fat mass, are involved in worsening glycaemic status: The Hiroshima study on glucose metabolism and cardiovascular diseases. Diabetes Obes Metab 2025; 27:3025-3035. [PMID: 40045548 DOI: 10.1111/dom.16307] [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: 11/21/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 05/04/2025]
Abstract
AIMS Insulin resistance in adipose tissue causes dysregulation of various adipokine levels and ectopic fat accumulation in other organs, such as the liver. This study investigated the effects of insulin resistance in adipose tissue and concomitant fatty liver on each stage of impaired glucose metabolism compared with visceral fat mass. MATERIALS AND METHODS This observational study included 3644 individuals who underwent two 75-g oral glucose tolerance tests at baseline and follow-up. Adipose insulin resistance index (Adipo-IR), lipid accumulation product (LAP) and fatty liver index (FLI) were used as indicators of insulin resistance in the adipose tissue, visceral fat mass and fatty liver, respectively. RESULTS Over a mean 2.9-year follow-up period, 463 (32.4%) individuals progressed from normoglycaemia to prediabetes, and 198 (10.6%) developed type 2 diabetes from prediabetes. Comparing the highest-to-lowest quartiles, baseline levels and changes in Adipo-IR were associated with an increased odds of progression from normoglycaemia to prediabetes (odds ratio [OR], 2.22; 95% CI, 1.36-3.65, OR, 2.70; 95% CI, 1.83-3.98, respectively) and from prediabetes to the onset of type 2 diabetes (OR, 2.02; 95% CI, 1.04-3.92, OR, 3.09; 95% CI, 1.84-4.19, respectively), after adjusting for possible confounders. Changes in FLI were associated with progression from prediabetes to type 2 diabetes. LAP did not affect the progression of impaired glucose metabolism. CONCLUSIONS Adipose tissue insulin resistance, rather than fat mass, is crucial in all stages of deterioration of glycaemic status. Fatty liver plays a decisive role in the eventual development of type 2 diabetes.
Collapse
Affiliation(s)
- Nobuo Sasaki
- Health Management and Promotion Center, Hiroshima Atomic Bomb Casualty Council, Hiroshima, Japan
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Ueno
- Health Management and Promotion Center, Hiroshima Atomic Bomb Casualty Council, Hiroshima, Japan
| | - Ryoji Ozono
- Department of General Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
2
|
Zhang N, Sun Q, Zhang J, Zhang R, Liu S, Zhao X, Ma J, Li X. Intrapancreatic adipocytes and beta cell dedifferentiation in human type 2 diabetes. Diabetologia 2025; 68:1242-1260. [PMID: 40072535 DOI: 10.1007/s00125-025-06392-9] [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: 10/04/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025]
Abstract
AIMS/HYPOTHESIS Fat deposition in the pancreas is implicated in beta cell dysfunction and the progress of type 2 diabetes. However, there is limited evidence to confirm the correlation and explore how pancreatic fat links with beta cell dysfunction in human type 2 diabetes. This study aimed to examine the spatial relationship between pancreatic fat and islets in human pancreases. METHODS Histological analysis of pancreatic specimens from 50 organ donors (15 with type 2 diabetes, 35 without) assessed pancreatic fat content variation among individuals with diabetes and its correlation with estimated beta cell mass and cell distribution within islets. Bioinformatic analysis of single-cell RNA-seq of 11 type 2 diabetic donors (from the Human Pancreatic Analysis Project database) explored the impact of high pancreatic fat content on beta cell gene expression and cell fate. Validation of bioinformatic results was performed with the above diabetic pancreases. RESULTS Pancreatic fat content was higher in individuals with type 2 diabetes (10.24% [3.29-13.89%] vs 0.74% [0.34-5.11%], p<0.001), negatively correlated with estimated beta cell mass (r=-0.675, p=0.006) and positively with alpha-to-beta cell ratio (r=0.608, p=0.016). Enrichment analysis indicated that in diabetic donors with higher pancreatic fat content, the expression of ALDH1A3, beta cell dedifferentiation marker, in both alpha and beta cells was significantly increased, and in beta cells, the expression of NPY decreased. Pseudotime analysis revealed beta cell dedifferentiation and transdifferentiation towards alpha cells in diabetic donors with higher pancreatic fat content, with decreased expression of genes related to beta cell maturation and function, including INSM1, MafA and NPY. Concurrently, pathways related to inflammation and immune response were activated. Histologically, pancreatic fat content correlated positively with the percentage of beta cells positive for aldehyde dehydrogenase 1 family member A3 (ALDH1A3) within the islets (r=0.594, p=0.020) and the ALDH1A3 positivity rate in beta cells (r=0.615, p=0.015). And the number of T cells adjacent to adipocytes was related to the distribution pattern of adipocytes and the dedifferentiation phenotype in islets. CONCLUSIONS/INTERPRETATION Higher pancreatic fat content was accompanied by increased beta cell dedifferentiation in the individuals with diabetes. Clusters of adipocytes significantly contribute to higher pancreatic fat content and immune cell recruitment. Overall, the interactions among adipocytes, immune cells and beta cells in the pancreas microenvironment might contribute to beta cell failure and dedifferentiation in type 2 diabetes.
Collapse
Affiliation(s)
- Na Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiman Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaxin Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruonan Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyi Liu
- Fudan University, Shanghai, China
| | - Xuelian Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Liu Y, Gang XK, Gao Y, Wang YX, Wang GX. Global status and trends in type 2 diabetes remission from 2002 to 2022: A bibliometric and visual analysis. Medicine (Baltimore) 2025; 104:e42257. [PMID: 40324218 PMCID: PMC12055079 DOI: 10.1097/md.0000000000042257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/23/2024] [Accepted: 11/28/2024] [Indexed: 05/07/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is estimated to impact 693 million individuals globally by 2045. Diabetes remission has the potential to slow disease progression, alleviate psychological burdens, minimize complications, and improve quality of life. We aimed to perform a bibliometric analysis of research on T2DM remission. We searched the Web of Science (WoS) database to identify relevant publications on T2DM remission during 2002 to 2022. Research trends and hotspots in T2DM remission were analyzed using Bibliometrix R and CiteSpace. The analysis considered various factors such as publication year, authors, journal, institution, country/region, themes, thematic evolution, keywords, and keyword bursts. The WoS search yielded 2254 articles. The annual scientific output has consistently increased. Lee was the most prolific author (48 papers). Obesity surgery was the leading journal (296 publications), while diabetes care had the highest h_index (43). The University of Copenhagen was the most active institution (116 papers). The most productive countries were the US (476), China (347), the UK (180), Italy (121), and Japan (90). The top 3 keywords were "bariatric surgery," "weight loss," and "remission." From 2013 to 2015, the usage of the term "medical therapy" significantly surged, lasting for 3 years. The term "GLP-I receptor agonists" also had a lasting burst. In the past 5 years, "weight loss" and "low-calorie diets" have emerged as prominent areas of research. This study analyzed the research trends and key factors in the field of type 2 diabetes mitigation through bibliometrics, providing important data support and a basis for decision-making for future research and public health policies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Kun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yuan Gao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ying-Xuan Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gui-Xia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
4
|
Taylor R. Aetiology of type 2 diabetes: an experimental medicine odyssey. Diabetologia 2025:10.1007/s00125-025-06428-0. [PMID: 40316731 DOI: 10.1007/s00125-025-06428-0] [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: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 05/04/2025]
Abstract
This review describes a prolonged research endeavour to test the twin cycle hypothesis that type 2 diabetes is caused by fat-induced dysfunction of the liver and pancreas, guided by the happenstance of clinical practice. Testing of the personal fat threshold hypothesis, that individuals exhibit different levels of tolerance to intra-organ fat accumulation, is also described. Both hypotheses predict that type 2 diabetes is potentially reversible by weight loss. The results of the Counterpoint study supported the twin cycle hypothesis, leading to a second study which determined that short-duration diabetes was more likely to remit following the 10-15 kg weight loss. It also confirmed that remission was durable over 6 months on an isoenergetic, normal diet. Subsequently, it was shown that weight loss caused an immediate decrease of pancreas fat only in people with type 2 diabetes and also that postprandial incretin spikes after bariatric surgery had no role in normalising fasting plasma glucose. DiRECT, a 2 year randomised controlled study, demonstrated clinical utility, observing functional beta cell capacity to return almost to normal over 12 months. A small group of participants regained weight and redeveloped type 2 diabetes, allowing observation that the underlying pathophysiological mechanisms during onset of diabetes were as postulated by the twin cycle hypothesis. Major clinical benefit was demonstrated after a further 3 year follow-up in routine care, halving the incidence of serious adverse effect compared with the standard treatment control group. In answer to the question of whether individuals have a personal fat threshold for tolerance of fat, stepwise weight loss in people with type 2 diabetes and BMI in the range 21-27 kg/m2 resulted in remission in 70%, with a wide range of fat thresholds. Type 2 diabetes can be regarded as a condition of homogenous aetiology in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Liang Q, Liu X, Xu X, Chen Z, Luo T, Su Y, Xie C. Molecular Mechanisms and Therapeutic Perspectives of Luteolin on Diabetes and Its Complications. Eur J Pharmacol 2025:177691. [PMID: 40311831 DOI: 10.1016/j.ejphar.2025.177691] [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: 03/10/2025] [Revised: 04/13/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Extensive preclinical studies have established luteolin, a flavonoid with potent antidiabetic activity, as a therapeutic candidate for preventing and managing various diabetic complications including cardiomyopathy, nephropathy, and osteopathy. This systematic review evaluates current evidence regarding luteolin's antidiabetic potential. AIM OF THE STUDY This study evaluates luteolin's efficacy in diabetes management through evidence synthesis, while critically assessing current research challenges and translational opportunities. METHODS A comprehensive literature search was conducted across Pubmed, Embase, Web of Science, and Google Scholar databases, encompassing articles published between 2000-2024. RESULTS Luteolin is a naturally occurring flavonoid that has strong antidiabetic properties. It regulates intestinal microenvironmental homeostasis, lipogenesis and catabolism, and the absorption of carbohydrates. It also modulates nine diabetic complications by reducing inflammation, oxidative stress, apoptosis, and autophagy. Luteolin's potential nutritional and physiological benefits notwithstanding, attention must be directed immediately to its bioavailability, innovative formulations, safety assessment, synergistic effects, and optimal dosage and time for supplementation. In particular, clinical studies are needed to validate efficacy and safety and provide a reliable scientific basis. CONCLUSION Luteolin may act as a pleiotropic molecule targeting multiple signaling cascades to exert antidiabetic bioactivity.
Collapse
Affiliation(s)
- Qingzhi Liang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Xiaoqin Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Xin Xu
- Department of Emergency, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Zhengtao Chen
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Ting Luo
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Yi Su
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Chunguang Xie
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 610072, China; Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
6
|
Fulda ES, Portas L, Harper C, Preiss D, Bennett D, Doherty A. Association of Daily Steps with Incident Non-Alcoholic Fatty Liver Disease: Evidence from the UK Biobank Cohort. Med Sci Sports Exerc 2025:00005768-990000000-00789. [PMID: 40279651 PMCID: PMC7617666 DOI: 10.1249/mss.0000000000003738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
PURPOSE Low physical activity has been shown to be associated with higher risk of non-alcoholic fatty liver disease (NAFLD). However, the strength and shape of this association are currently uncertain due to a reliance on self-reported physical activity measures. This report aims to investigate the relationship of median daily step count with NAFLD using accelerometer-derived step count from a large prospective cohort study. METHODS The wrist-worn accelerometer sub-study of the UK Biobank (N = ~100,000) was used to characterise median daily step count over a seven-day period. NAFLD cases were ascertained via record linkage with hospital inpatient data and death registers or by using a measure of liver fat from imaging. Cox proportional hazards models were employed to assess the association between step count and NAFLD, adjusting for age, sociodemographic, and lifestyle factors. Mediation analyses were conducted. RESULTS Among 91,031 participants (709,440 person-years of follow-up), there were 762 incident NAFLD cases. Higher step count was log-linearly and inversely associated with risk of NAFLD. A 1000-step increase (representing 10 minutes of walking) was associated with a 12% (95% CI: 10%-14%) lower hazard of NAFLD. When using imaging to identify NAFLD, a 1,000-step increase was associated with a 6% (95% CI: 6%-7%) lower risk. There was evidence for mediation by adiposity, accounting for 39% of the observed association. CONCLUSIONS Daily step count, a modifiable risk factor, is log-linearly and inversely associated with NAFLD. This association was only partially explained by adiposity. These findings from a large cohort study may have important implications for strategies to lower NAFLD risk.
Collapse
Affiliation(s)
- Evelynne S. Fulda
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Laura Portas
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Charlie Harper
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - David Preiss
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Aiden Doherty
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| |
Collapse
|
7
|
Li Y, Wei Z, Deng X. Reassessing β-Cell plasticity and weight loss Thresholds in Long-Standing type 2 diabetes remission. Diabetes Res Clin Pract 2025; 224:112198. [PMID: 40274106 DOI: 10.1016/j.diabres.2025.112198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Affiliation(s)
- Yadi Li
- Department of TCM, Deyang People's Hospital, Deyang, China.
| | - Zheng Wei
- Department of TCM, Deyang People's Hospital, Deyang, China
| | - Xinmin Deng
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Lee SY. Jejunoileal side-to-side anastomosis as a promising option for type 2 diabetes. World J Diabetes 2025; 16:103546. [PMID: 40236858 PMCID: PMC11947931 DOI: 10.4239/wjd.v16.i4.103546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
In this editorial, I discuss the article by Wang et al, published in the World Journal of Diabetes, which explores jejunoileal side-to-side anastomosis as a novel surgical intervention for type 2 diabetes mellitus (T2DM). T2DM, often associated with obesity, remains a global health challenge, as sustained remission is difficult to achieve with conventional pharmacological therapy. Jejunoileal anastomosis offers a promising alternative, particularly for patients with normal or relatively high body mass index, and addresses the unique challenges posed by diverse patient populations. This procedure preserves gastric anatomy while simultaneously improving metabolic parameters, such as glycemic control, lipid profiles, and pancreatic β-cell function. Unlike traditional metabolic surgeries that involve permanent anatomical alterations, this approach provides advantages such as reversibility, shorter operative times, and minimal nutritional complications, making it appealing to patients for whom conventional bariatric surgery is unsuitable. Advances in gut hormone physiology and incretin modulation support these findings. This innovative approach represents a potential paradigm shift in T2DM treatment, offering insights into the evolving role of surgical interventions in metabolic regulation. While early findings show promising diabetes remission rates and metabolic improvements at six months post-surgery, further studies with longer follow-up periods and broader patient cohorts are required.
Collapse
Affiliation(s)
- Sang Yeoup Lee
- Family Medicine and Biomedical Research Institute, Pusan National University Yangsan Hospital, Yangsan 50612, South Korea
- Medical Education, Pusan National University School of Medicine, Yangsan 50760, South Korea
| |
Collapse
|
9
|
Cheng AYY, Heine RJ, Del Prato S, Green JB, Thieu VT, Zeytinoglu M. Striving for early effective glycaemic and weight management in type 2 diabetes: A narrative review. Diabetes Obes Metab 2025; 27:1708-1718. [PMID: 39871817 PMCID: PMC11885087 DOI: 10.1111/dom.16206] [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: 10/07/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/29/2025]
Abstract
Despite the recognition by key guidelines that achieving early glycaemic control has important benefits in individuals with type 2 diabetes (T2D) and that addressing excess adiposity is one of the central components of comprehensive person-centred T2D care, a substantial proportion of individuals with T2D do not meet their metabolic treatment goals. Prior treatment paradigms were limited by important treatment-associated risks such as hypoglycaemia and body weight gain. Therefore, a more conservative, sequential approach to treatment was typically utilized. One potential consequence of this approach has been a missed opportunity to achieve a 'legacy effect', where early treatment to reach glycaemic targets is associated with enduring long-term benefits in T2D. Additionally, while previous treatment approaches have addressed core defects in T2D, including insulin resistance and β-cell function decline, they have been unable to address one of the underlying causal abnormalities-excess adiposity. Here, we review currently available evidence for the beneficial long-term effects of early glycaemic control and management of body weight in people with T2D and discuss potential next steps.
Collapse
|
10
|
Matsubayashi Y, Fujihara K, Khin L, Ferreira ED, Takabayashi S, Yamashita Y, Yamada T, Kodama S, Sone H. Association of changes in the type 2 diabetes and MASLD/related SLD status with risk of developing cardiovascular disease. Diabetes Obes Metab 2025; 27:2035-2043. [PMID: 39810604 PMCID: PMC11885070 DOI: 10.1111/dom.16196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND & AIMS This study assessed the association of remission of type 2 diabetes mellitus (DM) or metabolic dysfunction-associated steatotic liver disease (MASLD)/related SLD (r-SLD; MASLD with excessive alcohol intake) as defined by the fatty liver index with the risk of cardiovascular disease (CVD). METHODS Health examination data at baseline and after 2 years (2-Years) were extracted from a nationwide claims database in Japan. Among participants aged 18-72 years with at least 3 years of follow-up, 9345 participants with DM-associated MASLD/r-SLD and 71 932 participants with non-DM MASLD/r-SLD at baseline were included in the study. The participants were stratified by the achievement of remission of MASLD/r-SLD or DM at 2-Years. In each group after stratification, the risk of new-onset CVD during the observation period was analysed using multivariate Cox proportional hazards models. RESULTS During a median follow-up of 4.9 years (starting from 2-Years), 1368 cases of CVD were observed. The hazard ratio (95% confidence interval) for CVD was 0.50 (0.31-0.80) for participants with remission of DM, 0.65 (0.47-0.91) for participants with remission of MASLD/r-SLD, and 0.34 (0.15-0.77) for participants with remission of both DM and MASLD/r-SLD. Conversely, remission of MASLD/r-SLD was not linked to a reduced risk of CVD in participants with non-DM MASLD/r-SLD. CONCLUSION The association of MASLD/r-SLD remission with CVD risk differs greatly in the presence and absence of DM. In patients with DM-MASLD/r-SLD, MASLD/r-SLD remission can significantly reduce CVD risk similarly as remission of DM.
Collapse
Affiliation(s)
- Yasuhiro Matsubayashi
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Laymon Khin
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Efrem d'Àvila Ferreira
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Shizuka Takabayashi
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Yuko Yamashita
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Takaho Yamada
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Satoru Kodama
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| |
Collapse
|
11
|
Sattar N, Lee MMY. Estimating direct tissue effects versus weight loss effects of incretin-based drugs for obesity on various chronic conditions. Lancet Diabetes Endocrinol 2025; 13:347-354. [PMID: 39870097 DOI: 10.1016/s2213-8587(24)00363-2] [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: 09/17/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 01/29/2025]
Abstract
The extent to which newer, incretin-based drugs for obesity improve disease outcomes via weight loss versus the direct effects of these drugs is the subject of intense interest. Although reductions in major adverse cardiovascular events appear to be predominantly driven by the direct tissue effects of such drugs, the associated weight loss effects must be relevant to the benefits observed in other major outcomes, albeit to differing extents. In this Personal View, we draw on evidence to support that weight loss is at least partly responsible (albeit to differing extents) for the reported benefits of incretin-based drugs for obesity in people living with heart failure with preserved ejection fraction, hypertension, chronic kidney disease, and type 2 diabetes. Concurrently, we propose that drug-induced weight loss is largely responsible for the reported improvements in osteoarthritis, obstructive sleep apnoea, and metabolic dysfunction-associated steatohepatitis outcomes. However, more evidence is needed to solidify these observations, including, when possible, trials comparing the effects of incretin-based drugs for obesity with calorie-reduced diets on both outcomes and mechanistic pathways. Such evidence has implications for public health and the design of future trials of novel drugs for obesity.
Collapse
Affiliation(s)
- Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK.
| | - Matthew M Y Lee
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
12
|
Wong HJ, Lin NHY, Teo YH, Yeo BSY, Toh KZX, Teo YN, Chan MY, Yeo LLL, Poh KK, Kong WKF, Eng PC, Tan BYQ, Dalakoti M, Sia CH. Anti-diabetic effects of GLP-1 receptor agonists on obese and overweight patients across diabetes status, administration routes, treatment duration and baseline characteristics: A systematic review. Diabetes Obes Metab 2025; 27:1648-1659. [PMID: 39726212 DOI: 10.1111/dom.16136] [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: 08/13/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are increasingly used for anti-obesity indications. However, little is known of the comparative effect of GLP-1 RAs and their glycemic impact across the different routes of administration, diabetic statuses and durations of prescription. PubMed, EMBASE and CENTRAL were searched from inception to 13 February 2024. Only randomised controlled trials were included in this systematic review and meta-analysis. Adults aged above 18 years old, who were in the overweight/obesity range, with or without type 2 diabetes mellitus (T2DM) were included. Baseline characteristics and changes in glycated haemoglobin (HbA1c) and fasting plasma glucose (FPG) were obtained. GLP1-RAs demonstrated an overall reduction in HbA1c of -0.72% (95% confidence interval [CI] -0.79 to -0.65, p < 0.01) and in FPG of -1.00 mmol/L (95% CI -1.16 to -0.84, p < 0.01). HbA1c reduction in pre-DM patients was -0.44% (95% CI -0.54 to -0.18, p < 0.01). Patients who were followed up for more than a year experienced a smaller reduction of HbA1c. Meta-regression showed that the GLP-1 RAs are more efficacious at higher HbA1c and lower body mass index. Overall, GLP-1 RAs consistently led to a significant reduction in HbA1c at -0.72% and FPG at -1.00 mmol/L. These effects may be equally efficacious in pre-DM patients with obesity and those at lower BMI. With pre-DM and obesity being risk factors for metabolic syndrome, these findings may provide newer perspectives in expanding indications for GLP-1 RA initiation.
Collapse
Affiliation(s)
- Hon Jen Wong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Norman H Y Lin
- Department of Medicine, National University Hospital, Singapore
| | - Yao Hao Teo
- Department of Medicine, National University Hospital, Singapore
| | - Brian S Y Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Yao Neng Teo
- Department of Medicine, National University Hospital, Singapore
| | - Mark Y Chan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre Singapore, Singapore
| | - Leonard L L Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Neurology, Department of Medicine, National University Hospital, Singapore
| | - Kian Keong Poh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre Singapore, Singapore
| | - William K F Kong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre Singapore, Singapore
| | - Pei Chia Eng
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore
| | - Benjamin Y Q Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Neurology, Department of Medicine, National University Hospital, Singapore
| | - Mayank Dalakoti
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre Singapore, Singapore
| | - Ching-Hui Sia
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre Singapore, Singapore
| |
Collapse
|
13
|
Wu X, Huang Q, Ding Y, Cao Q, Jiang Y, Xu Y, Zhao Z, Xu M, Lu J, Wang T, Ning G, Wang W, Bi Y, Xu Y, Li M. Effect of insulin sensitivity, insulin secretion, and beta cell function on the remission of type 2 diabetes: A post hoc analysis of the IDEATE trial. Diabetes Obes Metab 2025; 27:1868-1877. [PMID: 39806566 DOI: 10.1111/dom.16180] [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: 10/09/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
AIMS To compare the probability of achieving diabetes remission in individuals with different phenotypes of insulin sensitivity, insulin secretion, and beta cell function and further detect the effects of diet, exercise, and lifestyle education intervention on these indexes. METHODS Three-hundred and one participants who had glycated haemoglobin (HbA1c) data at baseline and after intervention were included for this post hoc analysis. We used the multi-way analysis of variance to assess the differences between the diabetes remission and non-remission groups or between intervention groups in changes of the indexes of insulin sensitivity, insulin secretion, and beta cell function. Furthermore, logistic regression analysis was used to identify the association between the diabetes remission and baseline and change of each insulin index. RESULTS Participants with a higher disposition index (DI) or higher adaptation index at baseline were more likely to achieve diabetes remission. The diabetes remission group had a significantly greater increase in AUCc-pep0-30/AUCgluc0-30, DI, and adaptation index compared with the non-remission group, while there were no between-group differences in indexes of insulin sensitivity. Participants with greater increases in insulin secretion and beta cell function were more likely to achieve diabetes remission. Indexes of beta cell function improved in all intervention groups, while the diet intervention induced significant improvement compared with lifestyle education. CONCLUSIONS These findings supported the importance of aggressively implementing intensive lifestyle interventions for individuals with type 2 diabetes at an early stage of the disease, when beta cell function was not yet significantly impaired, to promote achieving diabetes remission.
Collapse
Affiliation(s)
- Xianglin Wu
- 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyue Huang
- Lifecycle Health Management Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Ding
- 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyu Cao
- 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youjin Jiang
- 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, Shanghai National Center for Translational Medicine, 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, Shanghai National Center for Translational Medicine, 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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, Shanghai National Center for Translational Medicine, 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - 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, Shanghai National Center for Translational Medicine, 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, Shanghai National Center for Translational Medicine, 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Xu
- 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lifecycle Health Management Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Digital Medicine Innovation Center, 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, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Popoviciu MS, Salmen T, Reurean-Pintilei D, Voiculescu V, Pantea Stoian A. SGLT-2i-A Useful Tool for Real-Life Metabolic and Body Weight Control in Type 2 Diabetes Mellitus Patients. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:548. [PMID: 40142359 PMCID: PMC11944101 DOI: 10.3390/medicina61030548] [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: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: Elevated blood sugar poses an increasingly significant challenge to healthcare systems worldwide. We aimed to assess the efficacy of the SGLT-2i class in achieving metabolic control in patients with T2DM within a real-world standard-of-care regimen. Material and Methods: A prospective analysis was conducted over 6 months including individuals receiving care in an outpatient department, with baseline assessments and follow-ups at 3 and 6 months. Results: A total of 280 patients were assessed, with a mean age of 63.69 ± 9.16, 53.9% of which were males, with a mean DM duration of 9.06 ± 5.64 years, and a DM duration varying from 6 months to 24 years. Discussion: Real-world evidence bridges the gap between guidelines and practice. It emphasizes the need to overcome clinical inertia in order to optimize patient outcomes and contributes to the body of evidence supporting the efficacy of fixed-dose SGLT-2i combinations in managing T2DM and associated comorbidities. Conclusions: We demonstrate the significant clinical and therapeutic impact of SGLT-2i in T2DM patients in a real-world setting. This class of medication not only positively influences glycemic and weight control but also reduces CV risk factors and visceral adiposity.
Collapse
Affiliation(s)
| | - Teodor Salmen
- Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Delia Reurean-Pintilei
- Department of Medical-Surgical and Complementary Sciences, Faculty of Medicine and Biological Sciences, “Ștefan cel Mare” University, 720229 Suceava, Romania
| | - Vlad Voiculescu
- Dermatology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Anca Pantea Stoian
- Diabetes, Nutrition and Metabolic Diseases Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
15
|
Liu Z, Feng N, Wang S, Liu Y, Wang J, Tan Y, Dong Y, Sun Z, Du X, Xu Y, Tao F, Zhong VW. Low-calorie diets and remission of type 2 diabetes in Chinese: phenotypic changes and individual variability. Nutr J 2025; 24:42. [PMID: 40087696 PMCID: PMC11908006 DOI: 10.1186/s12937-025-01101-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Chinese have distinct phenotypes of type 2 diabetes (T2D) and obesity compared with people of other ethnicities, but using low-calorie diets to achieve T2D remission has never been conducted in Chinese. This study aimed to assess if T2D remission can be achieved using low-calorie formula diet (LCFD) and low-calorie real food-based diet (LCRFD) in Chinese similarly to other populations and to identify determinants of individual variability in T2D remission. METHODS This 6-month intervention consisted of a 3-month isocaloric intensive weight loss phase (815-835 kcal/d) and a 3-month weight maintenance phase. Enrolled participants with T2D had BMI of 24-45 kg/m2 and HbA1c level of 6.5-12.0% (< 6.5% if on medication). Everyone stopped anti-diabetic drugs on day 1 and was assigned to receive LCFD (n = 21) or LCRFD (n = 20). RESULTS At 6 months, 29.3% of participants had ≥ 12 kg weight loss, 39.0% lost ≥ 10% weight, and 56.1% achieved T2D remission. MRI-derived liver and pancreatic fat decreased significantly. Significant improvement was also seen in insulin sensitivity, continuous glucose monitoring-derived metrics, and various other cardiometabolic risk factors but not arginine-induced insulin secretory response. There was no difference in all outcomes between LCFD and LCRFD. Compared with responders for T2D remission, nonresponders were more likely to be women, and had more fat mass, longer diabetes duration, poorer glycemic control, and lower beta-cell function. CONCLUSIONS T2D remission rate and weight loss amount following low-calorie diet intervention in Chinese people were comparable to those reported from other populations, although individual variability existed. LCFD and LCRFD were similarly effective. TRIAL REGISTRATION The trial was registered with ClinicalTrials.gov: NCT05472272.
Collapse
Affiliation(s)
- Zhenxiu Liu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200025, China
| | - Nannan Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building 227 South Chongqing Rd, Shanghai, China
| | - Sujing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building 227 South Chongqing Rd, Shanghai, China
| | - Yang Liu
- Department of Clinical Nutrition, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Wang
- Department of Chinese Medicine & Integrative Medicine, Shanghai Geriatric Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Tan
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200025, China
| | - Ying Dong
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building 227 South Chongqing Rd, Shanghai, China
| | - Zhewei Sun
- Department of Health and Behavioral Studies, Teachers College, Columbia University, New York, USA
| | - Xihao Du
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building 227 South Chongqing Rd, Shanghai, China
| | - Yaqing Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building 227 South Chongqing Rd, Shanghai, China
| | - Feng Tao
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200025, China
| | - Victor W Zhong
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building 227 South Chongqing Rd, Shanghai, China.
| |
Collapse
|
16
|
Whelehan G, Dirks ML, West S, Abdelrahman DR, Murton AJ, Finnigan TJA, Wall BT, Stephens FB. High-protein vegan and omnivorous diets improve peripheral insulin sensitivity to a similar extent in people with type 2 diabetes. Diabetes Obes Metab 2025; 27:1143-1152. [PMID: 39604044 PMCID: PMC11802395 DOI: 10.1111/dom.16100] [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: 10/10/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND High-protein diets have been recognized as a potential strategy in the nutritional management of type 2 diabetes (T2D). Mycoprotein is a high-fibre, high-protein food ingredient previously shown to improve acute glycaemic control. We determined whether incorporating mycoprotein into a high-protein vegan diet would improve glycaemic control to a greater extent than an isonitrogenous omnivorous diet in people with T2D. METHODS Seventeen adults (f = 5, age = 58.3 ± 8.3 years, BMI = 32.9 ± 4.7 kg∙m-2, HbA1c = 60 ± 15 mmol∙mol-1) with T2D were randomly allocated to a 5-week eucaloric high-protein (30% energy from protein) diet, either an omnivorous diet (OMNI; 70% protein from omnivorous sources) or an isonitrogenous, mycoprotein-rich, vegan diet (VEG; 50% protein from mycoprotein). Glycaemic control was assessed using a two-step hyperinsulinaemic-euglycaemic clamp (HEC) with D-[6,6-2H2] glucose infusion, a mixed-meal tolerance test (MMTT) and continuous glucose monitoring. RESULTS The rate of glucose disappearance (RdT), glucose disposal rate and endogenous glucose production, as well as postprandial time-course of blood glucose, serum insulin and C-peptide were assessed during the HEC and MMTT, respectively. Both groups had improved peripheral insulin sensitivity (intervention effect, p = 0.006; increased RdT/Insulin of 1.0 ± 0.6 and 1.0 ± 0.3 mg kg-1 min-1 in OMNI and VEG, respectively), HbA1c (intervention; p = 0.001) and glycaemic variability (intervention; p = 0.040; increased time in-range of 11.8 ± 9.3% and 23.3 ± 12.9% in OMNI and VEG). There were no improvements in hepatic insulin sensitivity or in postprandial blood glucose and serum C-peptide (p > 0.05) during the MMTT. CONCLUSIONS High-protein diets, whether predicated on vegan or omnivorous proteins, can improve glycaemic control by increasing peripheral insulin sensitivity in people with T2D.
Collapse
Affiliation(s)
- Gráinne Whelehan
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Marlou L. Dirks
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
- Human and Animal PhysiologyWageningen UniversityWageningenThe Netherlands
| | - Sam West
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Doaa R. Abdelrahman
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexasUSA
- Sealy Center of AgingUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Andrew J. Murton
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexasUSA
- Sealy Center of AgingUniversity of Texas Medical BranchGalvestonTexasUSA
| | | | - Benjamin T. Wall
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Francis B. Stephens
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| |
Collapse
|
17
|
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] [MESH Headings] [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
|
18
|
Rubino F, Cummings DE, Eckel RH, Cohen RV, Wilding JPH, Brown WA, Stanford FC, Batterham RL, Farooqi IS, Farpour-Lambert NJ, le Roux CW, Sattar N, Baur LA, Morrison KM, Misra A, Kadowaki T, Tham KW, Sumithran P, Garvey WT, Kirwan JP, Fernández-Real JM, Corkey BE, Toplak H, Kokkinos A, Kushner RF, Branca F, Valabhji J, Blüher M, Bornstein SR, Grill HJ, Ravussin E, Gregg E, Al Busaidi NB, Alfaris NF, Al Ozairi E, Carlsson LMS, Clément K, Després JP, Dixon JB, Galea G, Kaplan LM, Laferrère B, Laville M, Lim S, Luna Fuentes JR, Mooney VM, Nadglowski J, Urudinachi A, Olszanecka-Glinianowicz M, Pan A, Pattou F, Schauer PR, Tschöp MH, van der Merwe MT, Vettor R, Mingrone G. Definition and diagnostic criteria of clinical obesity. Lancet Diabetes Endocrinol 2025; 13:221-262. [PMID: 39824205 PMCID: PMC11870235 DOI: 10.1016/s2213-8587(24)00316-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 01/20/2025]
Abstract
Current BMI-based measures of obesity can both underestimate and overestimate adiposity and provide inadequate information about health at the individual level, which undermines medically-sound approaches to health care and policy. This Commission sought to define clinical obesity as a condition of illness that, akin to the notion of chronic disease in other medical specialties, directly results from the effect of excess adiposity on the function of organs and tissues. The specific aim of the Commission was to establish objective criteria for disease diagnosis, aiding clinical decision making and prioritisation of therapeutic interventions and public health strategies. To this end, a group of 58 experts—representing multiple medical specialties and countries—discussed available evidence and participated in a consensus development process. Among these commissioners were people with lived experience of obesity to ensure consideration of patients’ perspectives. The Commission defines obesity as a condition characterised by excess adiposity, with or without abnormal distribution or function of adipose tissue, and with causes that are multifactorial and still incompletely understood. We define clinical obesity as a chronic, systemic illness characterised by alterations in the function of tissues, organs, the entire individual, or a combination thereof, due to excess adiposity. Clinical obesity can lead to severe end-organ damage, causing life-altering and potentially life-threatening complications (eg, heart attack, stroke, and renal failure). We define preclinical obesity as a state of excess adiposity with preserved function of other tissues and organs and a varying, but generally increased, risk of developing clinical obesity and several other non-communicable diseases (eg, type 2 diabetes, cardiovascular disease, certain types of cancer, and mental disorders). Although the risk of mortality and obesity-associated diseases can rise as a continuum across increasing levels of fat mass, we differentiate between preclinical and clinical obesity (ie, health vs illness) for clinical and policy-related purposes. We recommend that BMI should be used only as a surrogate measure of health risk at a population level, for epidemiological studies, or for screening purposes, rather than as an individual measure of health. Excess adiposity should be confirmed by either direct measurement of body fat, where available, or at least one anthropometric criterion (eg, waist circumference, waist-to-hip ratio, or waist-to-height ratio) in addition to BMI, using validated methods and cutoff points appropriate to age, gender, and ethnicity. In people with very high BMI (ie, >40 kg/m2), however, excess adiposity can pragmatically be assumed, and no further confirmation is required. We also recommend that people with confirmed obesity status (ie, excess adiposity with or without abnormal organ or tissue function) should be assessed for clinical obesity. The diagnosis of clinical obesity requires one or both of the following main criteria: evidence of reduced organ or tissue function due to obesity (ie, signs, symptoms, or diagnostic tests showing abnormalities in the function of one or more tissue or organ system); or substantial, age-adjusted limitations of daily activities reflecting the specific effect of obesity on mobility, other basic activities of daily living (eg, bathing, dressing, toileting, continence, and eating), or both. People with clinical obesity should receive timely, evidence-based treatment, with the aim to induce improvement (or remission, when possible) of clinical manifestations of obesity and prevent progression to end-organ damage. People with preclinical obesity should undergo evidence-based health counselling, monitoring of their health status over time, and, when applicable, appropriate intervention to reduce risk of developing clinical obesity and other obesity-related diseases, as appropriate for the level of individual health risk. Policy makers and health authorities should ensure adequate and equitable access to available evidence-based treatments for individuals with clinical obesity, as appropriate for people with a chronic and potentially life-threatening illness. Public health strategies to reduce the incidence and prevalence of obesity at population levels must be based on current scientific evidence, rather than unproven assumptions that blame individual responsibility for the development of obesity. Weight-based bias and stigma are major obstacles in efforts to effectively prevent and treat obesity; health-care professionals and policy makers should receive proper training to address this important issue of obesity. All recommendations presented in this Commission have been agreed with the highest level of consensus among the commissioners (grade of agreement 90–100%) and have been endorsed by 76 organisations worldwide, including scientific societies and patient advocacy groups.
Collapse
Affiliation(s)
- Francesco Rubino
- Metabolic and Bariatric Surgery, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK; King's College Hospital, London, UK.
| | - David E Cummings
- University of Washington, Seattle, WA, USA; Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Robert H Eckel
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ricardo V Cohen
- Center for the Treatment of Obesity and Diabetes, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
| | - John P H Wilding
- Department of Cardiovascular and Metabolic Medicine, University of Liverpool, Liverpool, UK
| | - Wendy A Brown
- Monash University Department of Surgery, Central Clinical School, Alfred Health, Melbourne, VIC, Australia
| | - Fatima Cody Stanford
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rachel L Batterham
- International Medical Affairs, Eli Lilly, Basingstoke, UK; Diabetes and Endocrinology, University College London, London, UK
| | - I Sadaf Farooqi
- Institute of Metabolic Science and National Institute for Health and Care Research, Cambridge Biomedical Research Centre at Addenbrookes Hospital, Cambridge, UK
| | - Nathalie J Farpour-Lambert
- Obesity Prevention and Care Program, Department of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Carel W le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Louise A Baur
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Weight Management Services, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, Department of Pediatrics, McMaster University, Hamilton, ON, Canada; McMaster Children's Hospital, Hamilton, ON, Canada
| | - Anoop Misra
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India; National Diabetes Obesity and Cholesterol Foundation, New Delhi, India; Diabetes Foundation New Delhi, India
| | | | - Kwang Wei Tham
- Department of Endocrinology, Woodlands Health, National Healthcare Group, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Priya Sumithran
- Department of Surgery, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, Alfred Health, Melbourne, VIC, Australia
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John P Kirwan
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - José-Manuel Fernández-Real
- CIBER Pathophysiology of Obesity and Nutrition, Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain; Hospital Trueta of Girona and Institut d'Investigació Biomèdica de Girona, Girona, Spain
| | - Barbara E Corkey
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Hermann Toplak
- Division of Endocrinology and Diabetology, Department of Medicine, University of Graz, Graz, Austria
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert F Kushner
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francesco Branca
- Department of Nutrition and Food Safety, World Health Organization, Geneva, Switzerland
| | - Jonathan Valabhji
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK; Department of Diabetes and Endocrinology, Chelsea and Westminster Hospital National Health Service Foundation Trust, London, UK
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research of Helmholtz Munich, University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, Carl Gustav Carus University Hospital Dresden, Technical University Dresden, Dresden, Germany; School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Harvey J Grill
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Edward Gregg
- School of Population Health, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland; School of Public Health, Imperial College London, London, UK
| | - Noor B Al Busaidi
- National Diabetes and Endocrine Center, Royal Hospital, Muscat, Oman; Oman Diabetes Association, Muscat, Oman
| | - Nasreen F Alfaris
- Obesity Endocrine and Metabolism Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ebaa Al Ozairi
- Clinical Research Unit, Dasman Diabetes Institute, Dasman, Kuwait
| | - Lena M S Carlsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches, NutriOmics Research Group, INSERM, Sorbonne Université, Paris, France; Department of Nutrition, Pitié-Salpêtrière Hospital, Assistance Publique-Hospital of Paris, Paris, France
| | | | - John B Dixon
- Iverson Health Innovation Research institute, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Gauden Galea
- Regional Office for Europe, World Health Organization, Geneva, Switzerland
| | - Lee M Kaplan
- Section on Obesity Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Blandine Laferrère
- Division of Endocrinology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seoul, South Korea
| | | | - Vicki M Mooney
- European Coalition for people Living with Obesity, Dublin, Ireland
| | | | - Agbo Urudinachi
- Department of Community Health, Alex Ekwueme Federal University Teaching Hospital Abakaliki, Abakaliki, Nigeria
| | - Magdalena Olszanecka-Glinianowicz
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Science, Medical University of Silesia, Katowice, Poland
| | - An Pan
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Francois Pattou
- Translational Research for Diabetes, Lille University, Lille University Hospital, Inserm, Institut Pasteur Lille, Lille, France; Department of General and Endocrine Surgery, Lille University Hospital, Lille, France
| | | | - Matthias H Tschöp
- Helmholtz Munich, Munich, Germany; Technical University of Munich, Munich, Germany
| | - Maria T van der Merwe
- University of Pretoria, Pretoria, South Africa; Nectare Waterfall City Hospital, Midrand, South Africa
| | - Roberto Vettor
- Internal Medicine, Center for the Study and the Integrated Treatment of Obesity, Department of Medicine, University of Padova, Padua, Italy; Center for Metabolic and Nutrition Related Diseases,Humanitas Research Hospital, Milan, Italy
| | - Geltrude Mingrone
- Division of Diabetes & Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK; Catholic University of the Sacred Heart, Rome, Italy; University Polyclinic Foundation Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
19
|
Hanick CJ, Berg KJ, Garvey WT, Goss AM, Steger FL, Richman JS, Peterson CM. Study protocol, menu design, and rationale for a study testing the effects of a whole fruit-rich diet on glycemic control, liver fat, pancreatic fat, and cardiovascular health in adults with type 2 diabetes. Nutr Res 2025; 135:82-100. [PMID: 39978247 DOI: 10.1016/j.nutres.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/22/2025]
Abstract
Dietary interventions such as very-low-calorie diets and low-carbohydrate diets can improve glycemic control and even induce type 2 diabetes (T2D) remission. However, less is known about the effects of carbohydrate-rich whole foods, such as whole fruit, in people with T2D. Therefore, the aim of this pilot study is to investigate the effects of a whole fruit-rich diet on glycemic control, ectopic fat, and cardiovascular risk factors in adults with T2D. In this pilot study, sixteen adults aged 20 to 70 years with insulin-independent T2D for ≤6 years will complete a 12-week controlled-feeding intervention while maintaining their weight. During the ramp-up phase (weeks 1-4), participants progressively consume more whole fruit. During weeks 5 to 12, participants eat a fruit-rich Mediterranean diet providing 50% of calories as whole fruit (∼16.4 servings/day). All outcomes are measured at weeks 0, 4, and 12. The primary outcome is glycemic control, assessed hierarchically by whether participants achieve nondiabetic glucose concentrations without antihyperglycemic medications; the total dose of antihyperglycemic medications; mean glucose during a three-hour oral glucose tolerance test; and mean 24-hour glucose from continuous glucose monitoring. Secondary outcomes include intrahepatic lipid, pancreatic fat, blood pressure, heart rate, and serum lipids. We hypothesize that a fruit-rich diet will improve glycemic control, reduce the need for antihyperglycemic medications, decrease ectopic fat, and improve cardiovascular risk factors. This novel study will help determine the effects of a whole fruit-rich diet on glycemic control and liver fat and whether diabetes remission may be possible without losing weight. This study was registered at ClinicalTrials.gov (NCT03758742).
Collapse
Affiliation(s)
- Cody J Hanick
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kelly J Berg
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amy M Goss
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Felicia L Steger
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Endocrinology, Metabolism, & Genetics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Joshua S Richman
- Division of Gastrointestinal Surgery, UAB Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney M Peterson
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
20
|
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
|
21
|
Zeng H, Yu F, Hu H, Han Y, Ye R, Cao C. Nonlinear relationship between hepatic steatosis index and reversion to normal glucose regulation in Chinese adults with prediabetes. Sci Rep 2025; 15:4269. [PMID: 39905126 PMCID: PMC11794635 DOI: 10.1038/s41598-025-88314-8] [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: 10/15/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
The hepatic steatosis index (HSI) has been demonstrated to have a significant correlation with prediabetes and diabetes; however, its potential association with reversion to normal glucose regulation (NGR) from prediabetes remains insufficiently investigated. The primary objective of this study was to elucidate the relationship between HSI levels and the probability of reversion to NGR among Chinese adults with prediabetes. This retrospective cohort study utilized health examination data from 11,241 Chinese adults with prediabetes. Cox proportional hazard models were employed to calculate hazard ratios (HR) and 95% confidence intervals (CI) to assess the association between HSI levels and reversion to NGR. Additionally, a Cox proportional hazards regression model incorporating cubic spline functions and smooth curve fitting was utilized to determine any nonlinear relationships between HSI levels and reversion to NGR. The results of the multivariate analyses demonstrated a significant association between reduced HSI levels and an increased likelihood of reversion to NGR (HR = 0.92, 95%CI: 0.89-0.95, P < 0.0001). Furthermore, a nonlinear relationship was identified between HSI levels and the reversion to NGR, with a critical threshold at an HSI value of 43.08. Below this threshold, a strong negative association was observed, markedly enhancing the probability of returning to normoglycemic status (HR = 0.91, 95% CI: 0.88-0.94, P < 0.0001). This study reveals a negative, nonlinear correlation between HSI levels and the reversion to NGR in individuals with prediabetes. These findings highlight the essential role of effectively managing HSI as part of comprehensive prediabetes treatment strategies, which may significantly enhance the likelihood of achieving normoglycemic status.
Collapse
Affiliation(s)
- Haiyong Zeng
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, Guangdong, China
| | - Fei Yu
- Department of Rehabilitation, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Dapeng New District Nan'ao Hospital, No.3002, Sungang West Road, Futian District, Shenzhen, 518000, Guangdong Province, China
| | - Haofei Hu
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, Guangdong, China
| | - Yong Han
- Department of Emergency, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, Guangdong, China
| | - Ruixue Ye
- Department of Rehabilitation, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Futian District, Shenzhen, 518000, Guangdong Province, China.
| | - Changchun Cao
- Department of Rehabilitation, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Dapeng New District Nan'ao Hospital, No.3002, Sungang West Road, Futian District, Shenzhen, 518000, Guangdong Province, China.
| |
Collapse
|
22
|
Taylor R. The Twin Cycle Hypothesis of type 2 diabetes aetiology: From concept to national NHS programme. Exp Physiol 2025. [PMID: 39898429 DOI: 10.1113/ep092009] [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: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
The development of magnetic resonance methods for quantifying intra-organ metabolites has permitted advances in the understanding of fasting and post-prandial carbohydrate and lipid handling in people with and without type 2 diabetes. Insulin resistance in the liver was shown to be related to excess intra-organ fat and was able to be returned to normal by weight loss. The practical effect of having muscle insulin sensitivity in the lower part of the wide normal range resulted in the obligatory shunting of carbohydrates via de novo lipogenesis into saturated fat. These observations provided the basis for the Twin Cycle Hypothesis of the aetiology of type 2 diabetes. Subsequent studies on people with type 2 diabetes confirmed the postulated pathophysiological abnormalities and demonstrated their reversibility by dietary weight loss of 10-15 kg. Overall, the fundamental understanding of the mechanisms causing type 2 diabetes has bridged physiological and clinical perspectives. Large population-based randomised controlled trials confirmed the practical clinical application of the method of achieving substantial weight loss, and an NHS programme is now in place offering potential remission to people within 6 years of diagnosis.
Collapse
Affiliation(s)
- Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
23
|
Pan Z, Chen Q, Lin H, Huang W, Li J, Meng F, Zhong Z, Liu W, Li Z, Qin H, Huang B, Chen Y. Enhanced accuracy and stability in automated intra-pancreatic fat deposition monitoring of type 2 diabetes mellitus using Dixon MRI and deep learning. Abdom Radiol (NY) 2025:10.1007/s00261-025-04804-3. [PMID: 39841227 DOI: 10.1007/s00261-025-04804-3] [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: 09/11/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
PURPOSE Intra-pancreatic fat deposition (IPFD) is closely associated with the onset and progression of type 2 diabetes mellitus (T2DM). We aimed to develop an accurate and automated method for assessing IPFD on multi-echo Dixon MRI. MATERIALS AND METHODS In this retrospective study, 534 patients from two centers who underwent upper abdomen MRI and completed multi-echo and double-echo Dixon MRI were included. A pancreatic segmentation model was trained on double-echo Dixon water images using nnU-Net. Predicted masks were registered to the proton density fat fraction (PDFF) maps of the multi-echo Dixon sequence. Deep semantic segmentation feature-based radiomics (DSFR) and radiomics features were separately extracted on the PDFF maps and modeled using the support vector machine method with 5-fold cross-validation. The first deep learning radiomics (DLR) model was constructed to distinguish T2DM from non-diabetes and pre-diabetes by averaging the output scores of the DSFR and radiomics models. The second DLR model was then developed to distinguish pre-diabetes from non-diabetes. Two radiologist models were constructed based on the mean PDFF of three pancreatic regions of interest. RESULTS The mean Dice similarity coefficient for pancreas segmentation was 0.958 in the total test cohort. The AUCs of the DLR and two radiologist models in distinguishing T2DM from non-diabetes and pre-diabetes were 0.868, 0.760, and 0.782 in the training cohort, and 0.741, 0.724, and 0.653 in the external test cohort, respectively. For distinguishing pre-diabetes from non-diabetes, the AUCs were 0.881, 0.688, and 0.688 in the training cohort, which included data combined from both centers. Testing was not conducted due to limited pre-diabetic patients. Intraclass correlation coefficients between radiologists' pancreatic PDFF measurements were 0.800 and 0.699 at two centers, suggesting good and moderate reproducibility, respectively. CONCLUSION The DLR model demonstrated superior performance over radiologists, providing a more efficient, accurate and stable method for monitoring IPFD and predicting the risk of T2DM and pre-diabetes. This enables IPFD assessment to potentially serve as an early biomarker for T2DM, providing richer clinical information for disease progression and management.
Collapse
Affiliation(s)
- Zhongxian Pan
- Department of Radiology, Shenzhen Traditional Chinese Medicine Hospital (The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine), Shenzhen, China
| | - Qiuyi Chen
- Department of Radiology, Shenzhen Traditional Chinese Medicine Hospital (The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine), Shenzhen, China
| | - Haiwei Lin
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China
| | - Wensheng Huang
- Department of Radiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Junfeng Li
- Department of Radiology, Shenzhen Traditional Chinese Medicine Hospital (The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine), Shenzhen, China
| | - Fanqi Meng
- Department of Radiology, Shenzhen Traditional Chinese Medicine Hospital (The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine), Shenzhen, China
| | - Zhangnan Zhong
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China
| | - Wenxi Liu
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China
| | - Zhujing Li
- Department of Radiology, Shenzhen Traditional Chinese Medicine Hospital (The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine), Shenzhen, China
| | - Haodong Qin
- MR Research Collaboration, Siemens Healthineers, Shanghai, China
| | - Bingsheng Huang
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China.
| | - Yueyao Chen
- Department of Radiology, Shenzhen Traditional Chinese Medicine Hospital (The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine), Shenzhen, China.
| |
Collapse
|
24
|
Ye J, Wang JG, Liu RQ, Shi Q, Wang WX. Association between intra-pancreatic fat deposition and diseases of the exocrine pancreas: A narrative review. World J Gastroenterol 2025; 31:101180. [PMID: 39811515 PMCID: PMC11684206 DOI: 10.3748/wjg.v31.i2.101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/26/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Intrapancreatic fat deposition (IPFD) has garnered increasing attention in recent years. The prevalence of IPFD is relatively high and associated with factors such as obesity, age, and sex. However, the pathophysiological mechanisms underlying IPFD remain unclear, with several potential contributing factors, including oxidative stress, alterations in the gut microbiota, and hormonal imbalances. IPFD was found to be highly correlated with the occurrence and prognosis of exocrine pancreatic diseases. Although imaging techniques remain the primary diagnostic approach for IPFD, an expanding array of biomarkers and clinical scoring systems have been identified for screening purposes. Currently, effective treatments for IPFD are not available; however, existing medications, such as glucagon-like peptide-1 receptor agonists, and new therapeutic approaches explored in animal models have shown considerable potential for managing this disease. This paper reviews the pathogenesis of IPFD, its association with exocrine pancreatic diseases, and recent advancements in its diagnosis and treatment, emphasizing the significant clinical relevance of IPFD.
Collapse
Affiliation(s)
- Jing Ye
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Jian-Guo Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Rong-Qiang Liu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Qiao Shi
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Wei-Xing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| |
Collapse
|
25
|
Lorza-Gil E, Strauss OD, Ziegler E, Kansy K, Katschke MT, Rahimi G, Neuscheler D, Sandforth L, Sandforth A, Sancar G, Kaufmann B, Hartmann D, Singer S, Mihaljevic AL, Jumpertz-von Schwartzenberg R, Sbierski-Kind J, Müller TD, Birkenfeld AL, Gerst F. Incretin-responsive human pancreatic adipose tissue organoids: A functional model for fatty pancreas research. Mol Metab 2025; 91:102067. [PMID: 39549913 PMCID: PMC11625218 DOI: 10.1016/j.molmet.2024.102067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVE Infiltration of adipocytes into the pancreatic parenchyma has been linked to impaired insulin secretion in individuals with increased genetic risk of T2D and prediabetic conditions. However, the study of this ectopic fat depot has been limited by the lack of suitable in vitro models. METHODS Here, we developed a novel 3D model of functionally mature human pancreatic adipose tissue organoids by aggregating human pancreatic adipose tissue-derived stromal vascular fraction (SVF) cells into organoids and differentiating them over 19 days. RESULTS These organoids carry biological properties of the in situ pancreatic fat, presenting levels of adipogenic markers comparable to native pancreatic adipocytes and improved lipolytic and anti-lipolytic response compared to conventional 2D cultures. The organoids harbour a small population of immune cells, mimicking in vivo adipose environment. Furthermore, they express GIPR, allowing investigation of incretin effects in pancreatic fat. In accordance, GIP and the dual GLP1R/GIPR agonist tirzepatide stimulate lipolysis but had distinct effects on the expression of proinflammatory cytokines. CONCLUSIONS This novel adipose organoid model is a valuable tool to study the metabolic impact of incretin signalling in pancreatic adipose tissue, revealing potential therapeutic targets of incretins beyond islets. The donor-specific metabolic memory of these organoids enables examination of the pancreatic fat-islet crosstalk in a donor-related metabolic context.
Collapse
Affiliation(s)
- E Lorza-Gil
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.
| | - O D Strauss
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - E Ziegler
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - K Kansy
- German Center for Diabetes Research (DZD e.V.), Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - M-T Katschke
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany; M3 Research Center, University Hospital Tübingen, Tübingen, Germany
| | - G Rahimi
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - D Neuscheler
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - L Sandforth
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - A Sandforth
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - G Sancar
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - B Kaufmann
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - D Hartmann
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - S Singer
- Department of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - A L Mihaljevic
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - R Jumpertz-von Schwartzenberg
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - J Sbierski-Kind
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany; M3 Research Center, University Hospital Tübingen, Tübingen, Germany
| | - T D Müller
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany
| | - A L Birkenfeld
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - F Gerst
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
26
|
Janssen JAMJL. The Causal Role of Ectopic Fat Deposition in the Pathogenesis of Metabolic Syndrome. Int J Mol Sci 2024; 25:13238. [PMID: 39769002 PMCID: PMC11675790 DOI: 10.3390/ijms252413238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Consuming a "modern" Western diet and overnutrition may increase insulin secretion. Additionally, nutrition-mediated hyperinsulinemia is a major driver of ectopic fat deposition. The global prevalence of metabolic syndrome is high and growing. Within this context, people with congenital lipodystrophy often experience a severe form of metabolic syndrome. Evidence is increasingly supporting that subtle partial lipodystrophy plays an important role in the development of metabolic syndrome in the general population. In individuals in the general population with subtle partial lipodystrophy, as well as in those with congenital lipodystrophy, the subcutaneous adipose tissues are unable to accommodate surplus energy intake. In both conditions, (excess) fat is directed toward the liver, pancreas, and muscles, where it is deposited as ectopic fat, as this fat can no longer be stored in the "safe" subcutaneous fat depots. Ectopic fat depositions cause insulin resistance in the liver and muscles, as well as β-cell dysfunction in the pancreas. Support of a direct pathological role of ectopic fat deposition in this condition is further provided by the rapid normalization of hepatic insulin sensitivity and improvement in pancreatic β-cell function after marked reductions in ectopic fat depositions. Thus, ectopic fat deposition in the liver, pancreas, and muscles may play a causal role in the pathogenesis of metabolic syndrome even in the general population. As such, the prevention of ectopic fat deposition may reduce the risk of metabolic syndrome and mitigate its effects.
Collapse
Affiliation(s)
- Joseph A M J L Janssen
- Department of Internal Medicine, Erasmus Medical Center (Erasmus MC), Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
27
|
Skudder‐Hill L, Sequeira‐Bisson IR, Ko J, Poppitt SD, Petrov MS. The moderating effect of cardiometabolic factors on the association between hepatic and intrapancreatic fat. Obesity (Silver Spring) 2024; 32:2310-2320. [PMID: 39523209 PMCID: PMC11589540 DOI: 10.1002/oby.24154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Previous studies have investigated the association between hepatic fat and intrapancreatic fat deposition (IPFD); however, results have been inconclusive. The presence of cardiometabolic factors in certain subpopulations could explain this discrepancy. The aim of the present study was to use moderation analyses to determine the conditions under which hepatic fat is associated with IPFD. METHODS All participants underwent 3T abdominal magnetic resonance imaging (MRI) and spectroscopy. Hepatic fat and IPFD were manually quantified by independent raters. Moderation analyses were performed with adjustment for sex and ethnicity. RESULTS There were 367 participants included. Adjusted analyses of the overall cohort revealed that age, glycated hemoglobin (HbA1c), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides were significant moderators (p < 0.05) of the association between hepatic fat and IPFD. Ranges of significance included age < 61 years, HbA1c < 45 mmol/mol, LDL-C < 157 mg/dL, HDL-C > 36 mg/dL, and triglycerides < 203 mg/dL. CONCLUSIONS The association between hepatic fat and IPFD is generally present in young and middle-aged adults with good cardiometabolic health, whereas the link between the two fat depots becomes uncoupled in older adults or individuals with cardiometabolic risk factors.
Collapse
Affiliation(s)
| | - Ivana R. Sequeira‐Bisson
- Human Nutrition Unit, School of Biological SciencesUniversity of AucklandAucklandNew Zealand
- Riddet Centre of Research Excellence (CoRE) for Food and NutritionNew Zealand
| | - Juyeon Ko
- School of MedicineUniversity of AucklandAucklandNew Zealand
| | - Sally D. Poppitt
- School of MedicineUniversity of AucklandAucklandNew Zealand
- Human Nutrition Unit, School of Biological SciencesUniversity of AucklandAucklandNew Zealand
- Riddet Centre of Research Excellence (CoRE) for Food and NutritionNew Zealand
| | | |
Collapse
|
28
|
Dissanayake HA, Fernando DR, Nilaweera AI, Munasinghe TD, Kaushalya CMAU, Pulukkody MM, Katulanda P. Very-low-calorie diet-based intensive lifestyle intervention for remission of type 2 diabetes: Real-world experience in a South Asian population. Diabet Med 2024; 41:e15422. [PMID: 39118237 DOI: 10.1111/dme.15422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024]
Abstract
AIMS Very low-calorie diet (VLCD) can induce weight loss and diabetes remission (DR) amongst people with obesity and recent-onset type 2 diabetes (T2D). We aimed to determine the effectiveness and acceptability of VLCD in achieving DR amongst Sri Lankan adults with T2D. METHODS A retrospective analysis was conducted in a diabetes practice where VLCD-based Diabetes Remission Programme (VDRP) was offered for adults (>18 years) with T2D for <3 years and body mass index over 25 kg/m2. VLCD (~800 kcal/day, provided with/without diet replacement formula) was offered for 8-12 weeks, followed by gradual food reintroduction and exercise. DR was defined as HbA1c <6.5% at least 3 months after stopping glucose-lowering medications. RESULTS A total of 170 participants who enrolled in the VDRP (mean age 38.4 years [±11.1], men 68%, mean baseline HbA1c 86.9 [±18.1] mmol/mol (10.1 [±2.1]%), median duration of T2D 2 years [IQR 1-2]) and 87 (51%) of them followed the programme (attended at least one follow-up visit). Amongst the individuals who followed the VDRP, 40.2% achieved DR (35/87), compared with 2.4% (2/83) amongst those who did not follow the VDRP (aHR 9.3, 95% CI 2.2-16.4, p = 0.002). The proportion achieving normoglycaemia (HbA1c < 6.5%) but continued to take glucose-lowering medication was 20/87 among VDRP followers and 20/85 amongst VDRP non-followers. The commonest reasons for not following the VDRP were too restrictive dietary quantity (92%) and difficulties in finding recommended food items (67%). Majority (79%) would recommend VDRP to others. CONCLUSIONS VDRP is effective in achieving T2D remission amongst Sri Lankan adults with recently diagnosed T2D and obesity. Over half of the participants followed the programme and over 75% would recommend it to others, indicating good acceptability.
Collapse
Affiliation(s)
- H A Dissanayake
- Diabetes Research Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - D R Fernando
- Diabetes Research Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - A I Nilaweera
- Diabetes Research Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - T D Munasinghe
- Diabetes Research Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - C M A U Kaushalya
- Diabetes Research Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - M M Pulukkody
- Centre for Diabetes Endocrinology and Metabolism, Colombo, Sri Lanka
| | - P Katulanda
- Diabetes Research Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Centre for Diabetes Endocrinology and Metabolism, Colombo, Sri Lanka
| |
Collapse
|
29
|
Yamazaki H, Heni M, Wagner R, Fukuhara S, Grossman SR, Han S, Wu L, Streicher SA, Huang BZ. The Causal Effect of Intrapancreatic Fat Deposition on Acute and Chronic Pancreatitis: A Mendelian Randomization Study. Am J Gastroenterol 2024; 119:2540-2544. [PMID: 39162745 PMCID: PMC11617267 DOI: 10.14309/ajg.0000000000003048] [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: 06/05/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Recent associative studies have linked intrapancreatic fat deposition (IPFD) with risk of pancreatitis, but the causal relationship remains unclear. METHODS Using Mendelian randomization, we evaluated the causal association between genetically predicted IPFD and pancreatitis. This approach used genetic variants from genomewide association studies of IPFD (n = 25,617), acute pancreatitis (n = 6,787 cases/361,641 controls), and chronic pancreatitis (n = 3,875 cases/361,641 controls). RESULTS Genetically predicted IPFD was significantly associated with acute pancreatitis (odds ratio per 1-SD increase: 1.40 [95% CI: 1.12-1.76], P = 0.0032) and chronic pancreatitis (odds ratio: 1.64 [95% CI: 1.13-2.39], P = 0.0097). DISCUSSION Our findings support a causal role of IPFD in pancreatitis, suggesting that reducing IPFD could lower the risk of pancreatitis.
Collapse
Affiliation(s)
- Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for Innovative Research for Communities and Clinical Excellence (CiRC2LE), Fukushima Medical University, Fukushima, Japan
| | - Martin Heni
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, Ulm University, Ulm, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Róbert Wagner
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Shunichi Fukuhara
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for Innovative Research for Communities and Clinical Excellence (CiRC2LE), Fukushima Medical University, Fukushima, Japan
- Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Steven R. Grossman
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sihao Han
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Samantha A. Streicher
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Brian Z. Huang
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
30
|
Liu S, Wipf I, Joglekar A, Freshly A, Bovee CE, Kim L, Richtsmeier SL, Peachee S, Kopriva S, Vikram A, Ladiki DE, Ilerisoy F, Ilerisoy B, Sagona G, Jun C, Giedt M, Tootle TL, Ankrum J, Imai Y. Lipid droplet protein Perilipin 2 is critical for the regulation of insulin secretion through beta cell lipophagy and glucagon expression in pancreatic islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.17.624030. [PMID: 39605485 PMCID: PMC11601606 DOI: 10.1101/2024.11.17.624030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Knockdown (KD) of lipid droplet (LD) protein perilipin 2 (PLIN2) in beta cells impairs glucose-stimulated insulin secretion (GSIS) and mitochondrial function. Here, we addressed a pathway responsible for compromised mitochondrial integrity in PLIN2 KD beta cells. In PLIN2 KD human islets, mitochondria were fragmented in beta cells but not in alpha cells. Glucagon but not insulin level was elevated. While the formation of early LDs followed by fluorescent fatty acids (FA) analog Bodipy C12 (C12) was preserved, C12 accumulated in mitochondria over time in PLIN2 KD INS-1 cells. A lysosomal acid lipase inhibitor Lali2 prevented C12 transfer to mitochondria, mitochondrial fragmentation, and the impairment of GSIS. Direct interactions between LD-lysosome and lysosome-mitochondria were increased in PLIN2 KD INS-1 cells. Thus, FA released from LDs by microlipophagy cause mitochondrial changes and impair GSIS in PLIN2 KD beta cells. Interestingly, glucolipotoxic condition (GLT) caused C12 accumulation and mitochondrial fragmentation similar to PLIN2 KD in beta cells. Moreover, Lali2 reversed mitochondrial fragmentation and improved GSIS in human islets under GLT. In summary, PLIN2 regulates microlipophagy to prevent excess FA flux to mitochondria in beta cells. This pathway also contributes to GSIS impairment when LD pool expands under nutrient load in beta cells.
Collapse
|
31
|
Whelehan G, Bello O, Hakim O, Ladwa M, Umpleby AM, Amiel SA, Bodicoat DH, Goff LM. Ethnic differences in the relationship between ectopic fat deposition and insulin sensitivity in Black African and White European men across a spectrum of glucose tolerance. Diabetes Obes Metab 2024; 26:5211-5221. [PMID: 39149769 DOI: 10.1111/dom.15867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024]
Abstract
AIM To examine the hypothesis that there would be ethnic differences in the relationship between ectopic fat and tissue-specific insulin resistance (IR) across a spectrum of glucose tolerance in Black African (BA) and White European (WE) men. MATERIALS AND METHODS Fifty-three WE men (23/10/20 normal glucose tolerance [NGT]/impaired glucose tolerance [IGT]/type 2 diabetes [T2D]) and 48 BA men (20/10/18, respectively) underwent a two-step hyperinsulinaemic-euglycaemic clamp with infusion of D-[6,6-2H2]-glucose and [2H5]-glycerol to assess hepatic, peripheral and adipose tissue IR. Magnetic resonance imaging was used to measure subcutaneous adipose tissue, visceral adipose tissue (VAT) and intrahepatic lipid (IHL). Associations between ectopic fat and IR were assessed using linear regression models. RESULTS There were no differences in tissue-specific IR between ethnic groups at any stage of glucose tolerance. VAT level was consistently lower in the BA population; NGT (p = 0.013), IGT (p = 0.006) and T2D (p = 0.015). IHL was also lower in the BA compared with the WE men (p = 0.013). VAT and IHL levels were significantly associated with hepatic IR in the BA population (p = 0.001) and with peripheral IR in the WE population (p = 0.027). CONCLUSIONS The present study suggests that BA and WE men exhibit the same degree of IR across a glucose tolerance continuum, but with lower VAT and IHL levels in the BA population, suggesting that IR may be driven by a mechanism other than increased ectopic fat accumulation in BA men.
Collapse
Affiliation(s)
- Gráinne Whelehan
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, UK
| | - Oluwatoyosi Bello
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Olah Hakim
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Meera Ladwa
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - A Margot Umpleby
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Stephanie A Amiel
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | | - Louise M Goff
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, UK
| |
Collapse
|
32
|
Bergman BC, Zemski Berry K, Garfield A, Keller A, Zarini S, Bowen S, McKenna C, Kahn D, Pavelka J, Macias E, Uhlson C, Johnson C, Russ HA, Viesi CH, Seldin M, Liu C, Doliba N, Schoen J, Rothchild K, Hazel K, Naji A. Human peripancreatic adipose tissue paracrine signaling impacts insulin secretion, blood flow, and gene transcription. J Clin Endocrinol Metab 2024:dgae767. [PMID: 39484843 DOI: 10.1210/clinem/dgae767] [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: 05/13/2024] [Revised: 08/14/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024]
Abstract
CONTEXT Adipose tissue accumulation around non-adipose tissues is associated with obesity and metabolic disease. One relatively unstudied depot is peripancreatic adipose tissue (PAT) that accumulates in obesity and insulin resistance and may impact beta cell function. Pancreatic lipid accumulation and PAT content are negatively related to metabolic outcomes in humans, but these studies are limited by the inability to pursue mechanisms. OBJECTIVE We obtained PAT from human donors through the Human Pancreas Analysis Program to evaluate differences in paracrine signaling compared to subcutaneous adipose tissue (SAT), as well as effects of the PAT secretome on aortic vasodilation, human islet insulin secretion, and gene transcription using RNAseq. RESULTS PAT had greater secretion of IFN-γ and most inflammatory eicosanoids compared to SAT. Secretion of adipokines negatively related to metabolic health were also increased in PAT compared to SAT. We found no overall effects of PAT compared to SAT on human islet insulin secretion, however, insulin secretion was suppressed after PAT exposure from men compared to women. Vasodilation was significantly dampened by PAT conditioned media, an effect explained almost completely by PAT from men and not women. Islets treated with PAT showed selective changes in lipid metabolism pathways while SAT altered cellular signaling and growth. RNAseq analysis showed changes in islet gene transcription impacted by PAT compared to SAT, with the biggest changes found between PAT based on sex. CONCLUSION The PAT secretome is metabolically negative compared to SAT, and impacts islet insulin secretion, blood flow, and gene transcription in a sex dependent manner.
Collapse
Affiliation(s)
- Bryan C Bergman
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karin Zemski Berry
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amanda Garfield
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amy Keller
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Simona Zarini
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sophia Bowen
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Colleen McKenna
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Darcy Kahn
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jay Pavelka
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily Macias
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charis Uhlson
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chris Johnson
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Holger A Russ
- College of Medicine, Department of Pharmacology and Therapeutics, University of Florida USA
- Diabetes Institute, University of Florida USA
| | - Carlos H Viesi
- Department of Biological Chemistry and the Center for Epigenetics and Metabolism, University of California, Irvine, CA, USA
| | - Marcus Seldin
- Department of Biological Chemistry and the Center for Epigenetics and Metabolism, University of California, Irvine, CA, USA
| | - Chengyang Liu
- University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Nicolai Doliba
- University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Jonathan Schoen
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin Rothchild
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kweku Hazel
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ali Naji
- University of Pennsylvania Medical Center, Philadelphia, PA, USA
| |
Collapse
|
33
|
Iglesies-Grau J, Dionne V, Latour É, Gayda M, Besnier F, Gagnon D, Debray A, Gagnon C, Tessier AJ, Paradis A, Klai C, Martin N, Pelletier V, Simard F, Nigam A, L’Allier PL, Juneau M, Bouabdallaoui N, Bherer L. Cardiac Rehabilitation for Prediabetes and Metabolic Syndrome Remission: Impact of Ultraprocessed Food-Intake Reduction and Time-Restricted Eating in the DIABEPIC-1 Study. CJC Open 2024; 6:1411-1421. [PMID: 39582705 PMCID: PMC11583856 DOI: 10.1016/j.cjco.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/29/2024] [Indexed: 11/26/2024] Open
Abstract
Background Cardiac rehabilitation programs provide a valuable opportunity to promote the adoption of healthy lifestyle behaviors in patients with atherosclerotic cardiovascular diseases (ASCVDs) and metabolic comorbidities, including metabolic syndrome and prediabetes. However, strategies to reverse these conditions remain to be explored. The DIABEPIC-1 study aimed to assess the feasibility of an enhanced 6-month cardiac rehabilitation program for patients with ASCVD while investigating prediabetes and metabolic syndrome remission. Methods The study combined exercise training with a comprehensive nutritional intervention, emphasizing reduction in intake of ultraprocessed foods, adoption of a Mediterranean diet, and implementation of time-restricted eating. Baseline, 3-month, and 6-month assessments included segmental body-composition measurements, blood analysis, maximal exercise testing, nutritional diaries recorded with the Keenoa AI app, and lifestyle questionnaires. Remission criteria included a return to an HbA1c level of < 5.7%, and < 3 National Cholesterol Education Program Adult Treatment Panel III (NCEP/ATP-III) criteria for prediabetes and metabolic syndrome, respectively. Results A total of 36 participants were recruited. The study demonstrated completion rates of 94.4% at 3 months, and 88.9% at 6 months, and a mean compliance rate of 92.5% for planned clinical appointments. Significant reductions in waist circumference (-9.2 cm, P < 0.001) and weight (-8.0 kg, P < 0.001) were observed. Improvement in glycemic and lipid profiles, insulin-resistance marker levels, and liver health were noted. Participants enhanced their cardiorespiratory fitness, reduced their consumption of ultraprocessed food, and increased their adherence to the Mediterranean diet and time-restricted eating. Notably, 50% achieved prediabetes remission, and 70% with metabolic syndrome at baseline achieved remission. Conclusions The study demonstrates the possibility of enhancing cardiac rehabilitation with an intensive nutritional intervention, yielding clinically significant outcomes, including remission of key risk factors in a substantial number of ASCVD patients. Clinical Trial Registration ClinicalTrials.gov, NCT05459987.
Collapse
Affiliation(s)
- Josep Iglesies-Grau
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Valérie Dionne
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Élise Latour
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - Mathieu Gayda
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Florent Besnier
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Daniel Gagnon
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- School of Kinesiology and Exercise Science, Université de Montréal, Montréal, Québec, Canada
| | - Amélie Debray
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- School of Kinesiology and Exercise Science, Université de Montréal, Montréal, Québec, Canada
| | - Christine Gagnon
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - Anne-Julie Tessier
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - Andréanne Paradis
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - Chiheb Klai
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - Nicolas Martin
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - Véronique Pelletier
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
| | - François Simard
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Anil Nigam
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Philippe L. L’Allier
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Martin Juneau
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Nadia Bouabdallaoui
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Louis Bherer
- Research Center and Centre ÉPIC, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
- Research Center, Institut Universitaire de Gériatrie de Montréal, Montréal, Québec, Canada
| |
Collapse
|
34
|
Zhang Z, Bao K, Liu J, Jiang C, Ji C, Han J, Zhou G, Liu X, Liu T, Gao J, Wang X, Yan H. Acupuncture-assisted lifestyle intervention improve the metabolic status and spontaneous brain activity of type 2 diabetes Mellitus patients: a randomized, clinical trial. Diabetol Metab Syndr 2024; 16:255. [PMID: 39468590 PMCID: PMC11514942 DOI: 10.1186/s13098-024-01489-4] [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: 07/05/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Aggressive weight management in patients with type 2 diabetes mellitus has demonstrated numerous metabolic advantages, however, existing therapies for weight control have not reached satisfactory results. This study aimed to evaluate the efficacy and safety of acupuncture in the weight management of type 2 diabetes mellitus (T2DM) patients by using a randomized, sham-controlled clinical trial design. METHODS In this single-blind randomized clinical trial, 102 overweight adult T2DM patients were randomized into two groups. The control group receives diet, exercise, and sham acupuncture intervention, whereas the acupuncture group receives diet and exercise and acupuncture intervention, both for 1 month. Body weight and other anthropometric and laboratory indices were assessed at baseline and endpoint, meanwhile, the body fat content and spontaneous brain activity were measured by functional magnetic resonance imaging(fMRI)as the exploratory outcomes. RESULTS No significant difference was observed between the studied parameters at the baseline. The body weight and BMI were significantly reduced both in the control and acupuncture groups after intervention, without statistical difference between the two groups. What's interesting is that compared to the control, the acupuncture group displayed a greater improvement in central fat tissue. It notes that the acupuncture group achieved significant liver fat content reduction than the sham acupuncture group. At the same time, the spontaneous brain activity in the occipital lobe and parietal lobe significantly increased in the acupuncture group. CONCLUSION One month of acupuncture treatment preferentially improved ectopic fat deposition and was accompanied by changes in brain activity compared with the control group, even before significant changes in total body weight had occurred. further studies of longer duration are necessary for validation. TRIAL REGISTRATION The protocol of this clinical trial is registered at the Acupuncture-Moxibustion Clinical Trial Registry (AMCTR, http://www.acmctr.org/ , No. AMCTR-IOR-20000341).
Collapse
Affiliation(s)
- Zhitian Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Kaixuan Bao
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Jiaojiao Liu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chunfei Jiang
- Department of traditional Chinese medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chunyan Ji
- Department of Nutrition, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiawei Han
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 201203, China
| | - Guo Zhou
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Xiaoyu Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Tiemin Liu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, 200433, China
- School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Jian Gao
- Department of Nutrition, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Xiangyu Wang
- Department of traditional Chinese medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Fudan Zhangjiang Institute, Shanghai, 201203, China.
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Fudan Institute for Metabolic Diseases, Shanghai, 200032, China.
- Department of Endocrinology and Metabolism, Wusong Branch of Zhongshan Hospital, Fudan University, Shanghai, 200940, China.
| |
Collapse
|
35
|
Lyngbæk MPP, Legaard GE, Nielsen NS, Durrer C, Almdal TP, Lund MAV, Liebetrau B, Ewertsen C, Lauridsen C, Solomon TPJ, Karstoft K, Pedersen BK, Ried-Larsen M. Effects of caloric restriction with different doses of exercise on fat loss in people living with type 2 diabetes: A secondary analysis of the DOSE-EX randomized clinical trial. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 14:100999. [PMID: 39427878 PMCID: PMC11964559 DOI: 10.1016/j.jshs.2024.100999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/02/2024] [Accepted: 06/29/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Fat loss mainly conveys the benefits of caloric restriction for people living with type 2 diabetes. The literature is equivocal regarding whether exercise facilitates fat loss during caloric restriction. This analysis aimed to assess the dose-response effects of exercise in combination with a caloric restriction on fat mass (FM) and FM percentage (FM%) in persons with diagnosed type 2 diabetes. METHODS In this secondary analysis of a 4-armed randomized trial, 82 persons living with type 2 diabetes were randomly allocated to the control group (CON) (n = 21), diet control (DCON) (25% caloric restriction; n = 20), diet control and exercise 3 times per week (MED) (n = 20), or diet control and exercise 6 times per week (HED) (n = 21) for 16 weeks. The primary analysis was the change in FM% points. Secondary analyses included fat-free mass and visceral adipose tissue (VAT) volume (cm3). RESULTS FM% decreased compared to CON by a mean difference of -3.5% (95% confidence interval (95%CI): -5.6% to -1.4%), -6.3% (95%CI: -8.4% to -4.1%), and -8.0% (95%CI: -10.2% to -5.8%) for DCON, MED, and HED, respectively. Compared to DCON, MED, and HED decreased FM% by -2.8% (95%CI: -4.9% to -0.7%) and -4.5% (95%CI: -6.6% to -2.4%), respectively. The difference in FM% between HED and MED was -1.8% (95%CI: -3.9% to 0.4%). DCON and MED decreased fat-free mass compared to CON, whereas HED preserved fat-free mass (-0.2%; 95%CI: -2.0% to 1.7%). Compared to CON, VAT volume decreased by -666.0 cm3 (95%CI: -912.8 cm3 to -385.1 cm3), -1264.0 cm3 (95%CI: -1679.6 cm3 to -655.9 cm3), and -1786.4 cm3 (95%CI: -2264.6 cm3 to -1321.2 cm3) more for DCON, MED, and HED, respectively. HED decreased VAT volume more than DCON (-1120.4 cm3; 95%CI: -1746.6 cm3 to -639.4 cm3) while the remaining comparisons did not reveal any differences. CONCLUSION All interventions were superior in reducing FM% compared to standard care. Adding exercise to a caloric restriction was superior in reducing FM% compared to a caloric restriction alone.
Collapse
Affiliation(s)
- Mark P P Lyngbæk
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Grit E Legaard
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Nina S Nielsen
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Cody Durrer
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Thomas P Almdal
- Department of Endocrinology, University of Copenhagen-Rigshospitalet, Copenhagen 2100, Denmark; Department of Immunology & Microbiology, University of Copenhagen, Copenhagen 2200, Denmark
| | - Morten Asp Vonsild Lund
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Cardiology, University Hospital Copenhagen-Rigshospitalet, Copenhagen 2100, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Holbæk Hospital, Holbæk 4300, Denmark
| | - Benedikte Liebetrau
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Caroline Ewertsen
- Department of Radiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Carsten Lauridsen
- Department of Radiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark; Bachelor's Degree Programme in Radiography, Copenhagen University College, Copenhagen 1799, Denmark
| | | | - Kristian Karstoft
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark; Department of Clinical Pharmacology, Bispebjerg-Frederiksberg Hospital, University of Copenhagen, Copenhagen 2400, Denmark
| | - Bente K Pedersen
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark
| | - Mathias Ried-Larsen
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen 2100, Denmark.
| |
Collapse
|
36
|
Karmacharya B, Sapkota S, Rai P, Nikolaou C, Kasti R, Bhattarai J, Maharjan R, Shrestha A, Shrestha A, Bhattarai B, Leeds AR, McIntosh A, Lean MEJ. A service evaluation of weight management for glycaemic control and remission of type 2 diabetes using traditional food in Nepal (Ho-DIRECT NEPAL): a single-arm trial. THE LANCET REGIONAL HEALTH. SOUTHEAST ASIA 2024; 29:100465. [PMID: 39253354 PMCID: PMC11381750 DOI: 10.1016/j.lansea.2024.100465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/06/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024]
Abstract
Background Remission of early type 2 diabetes (T2D) is possible; however, diet programmes proven effective are unaffordable in many southeast Asian populations where T2D is more frequent and more aggressive at lower body weight and younger age. We evaluate an entirely food-based service. Methods This study employed a single-arm intervention and follow-up design for intervention evaluation in existing hospital people with T2D of under 5 years known duration. Individuals attending a diabetes clinic in Kathmandu with early T2D (<5 years) aged 30-70 years, BMI ≥23 kg/m2, were offered a low-cost nutritionally complete diet-programme, using traditional Nepali foods to provide 8-weeks ∼850 kcal/day weight loss induction, and then weight maintenance. The participants received 4-weekly dietetic appointments (30-45 min) and verbo-pictorial leaflets using household measures. Glucose-lowering medications (49/70 at baseline) were stopped at baseline or soon after. The study was registered as ISRCTN10671396, testing a traditional food-based intervention for weight loss and T2D remission. Findings For 70 individuals (45 female) invited between March 19, 2022 and September 19, 2023, baseline mean (SD) age was 48.6 (9.9) years, bodyweight 74.6 (9.5) kg, BMI 29.7 (3.6) kg/m2, known diabetes duration 2.5 (1.9) years, HbA1c on treatment 8.1 (1.6) %. At 12, 24 and 52 weeks respectively, evaluating n = 44, 46, 45, bodyweight was 70.1 (8.5), 69.8 (8.9), 70.0 (8.8) kg, HbA1c 6.8 (0.9), 6.9 (1.5), 7.1 (1.3) %; HbA1c <6.5% was recorded for 46%, 48% and 36% and remission of T2D (HbA1c <6.5% off medication >3 months) in 43%, 39% and 29%. The main reported adherence barriers were fears of weakness, hunger, and inconvenience during travel. Incentives were ease of the diet, reduced doses and costs of medications, and improved appearance. Interpretation Traditional food-based weight management can valuably improve control, reduce medication needs, and generate remissions of established T2D, but adherence barriers must be overcome to optimise outcomes. Funding All Saints Educational Trust, England.
Collapse
Affiliation(s)
- Biraj Karmacharya
- Department of Public Health and Community Programs, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | - Sujata Sapkota
- Manmohan Memorial Institute of Health Sciences, Kathmandu, Nepal
- Institute for Implementation Science and Health, Kathmandu, Nepal
| | - Prasanna Rai
- Department of Public Health and Community Programs, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | | | - Roshan Kasti
- Department of Public Health and Community Programs, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | | | - Rashmi Maharjan
- Department of Public Health and Community Programs, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
- Department of Epidemiology, University of Washington, WA, USA
| | - Abha Shrestha
- Department of Public Health and Community Programs, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
- Department of Community Medicine, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | - Archana Shrestha
- Department of Public Health and Community Programs, Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
- Institute for Implementation Science and Health, Kathmandu, Nepal
- Center of Methods for Implementation and Prevention Science, Yale School of Public Health, New Haven, CT, USA
| | | | - Anthony R Leeds
- Department of Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Alasdair McIntosh
- Robertson Centre for Biostatistics, School of Health and Wellbeing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Michael E J Lean
- Department of Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
37
|
Anyiam O, Abdul Rashid RS, Bhatti A, Khan-Madni S, Ogunyemi O, Ardavani A, Idris I. A Systematic Review and Meta-Analysis of the Effect of Caloric Restriction on Skeletal Muscle Mass in Individuals with, and without, Type 2 Diabetes. Nutrients 2024; 16:3328. [PMID: 39408294 PMCID: PMC11479040 DOI: 10.3390/nu16193328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Severe caloric restriction interventions (such as very-low-calorie diets) are effective for inducing significant weight loss and remission of type 2 diabetes (T2DM). However, suggestions of associated significant muscle mass (MM) loss create apprehension regarding their widespread use. We conducted a systematic review and meta-analysis to provide a quantitative assessment of their effect on measures of MM in individuals with, or without, T2DM. METHODS EMBASE, Medline, Pubmed, CINAHL, CENTRAL and Google Scholar were systematically searched for studies involving caloric restriction interventions up to 900 kilocalories per day reporting any measure of MM, in addition to fat mass (FM) or body weight (BW). RESULTS Forty-nine studies were eligible for inclusion, involving 4785 participants. Individuals with T2DM experienced significant reductions in MM (WMD -2.88 kg, 95% CI: -3.54, -2.22; p < 0.0001), although this was significantly less than the reduction in FM (WMD -7.62 kg, 95% CI: -10.87, -4.37; p < 0.0001). A similar pattern was observed across studies involving individuals without T2DM. MM constituted approximately 25.5% of overall weight loss in individuals with T2DM, and 27.5% in individuals without T2DM. Subgroup analysis paradoxically revealed greater BW and FM reductions with less restrictive interventions. CONCLUSIONS Our review suggests that caloric restriction interventions up to 900 kilocalories per day are associated with a significant reduction in MM, albeit in the context of a significantly greater reduction in FM. Furthermore, MM constituted approximately a quarter of the total weight loss. Finally, our data support the use of less restrictive interventions, which appear to be more beneficial for BW and FM loss.
Collapse
Affiliation(s)
- Oluwaseun Anyiam
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK
- Department of Endocrinology and Diabetes, University Hospitals of Derby and Burton NHS Foundation Trust, Derby DE22 3NE, UK
| | | | - Aniqah Bhatti
- Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
| | - Saif Khan-Madni
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Olakunmi Ogunyemi
- Department of Acute Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby DE22 3NE, UK
- Nuffield Department of Population Health, University of Oxford, Oxford OX1 2JD, UK
| | - Arash Ardavani
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK
- Department of Endocrinology and Diabetes, University Hospitals of Derby and Burton NHS Foundation Trust, Derby DE22 3NE, UK
| | - Iskandar Idris
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK
- Department of Endocrinology and Diabetes, University Hospitals of Derby and Burton NHS Foundation Trust, Derby DE22 3NE, UK
| |
Collapse
|
38
|
Shin S, Chang Y, Ryu S. Sex-specific association between carbohydrate antigen 19-9 and incident type 2 diabetes. Sci Rep 2024; 14:22506. [PMID: 39341838 PMCID: PMC11439046 DOI: 10.1038/s41598-024-73404-w] [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: 06/10/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Carbohydrate antigen 19-9 (CA19-9) levels are associated with glycemic control, insulin resistance, and chronic complications in patients with type 2 diabetes (T2D). Women generally show higher CA19-9 levels despite a greater T2D prevalence in men. We evaluated the sex-specific longitudinal associations between CA19-9 levels and T2D incidence. Korean adults (n = 329,380) without previous cancer or T2D were categorized into four groups based on their CA19-9 levels. The study end point was the development of incident T2D during follow-up. Cox proportional hazards models were used to estimate hazard ratios (HR) according to CA19-9 levels. During a median follow-up of 6.1 years (3.3-9.3 years), the incidence rates of T2D were 9.9 per 1,000 person-years in men and 3.6 per 1,000 person-years in women. In the time-dependent analysis, adjusted HRs (95% confidence intervals) for incident T2D comparing CA19-9 levels of 10.0-19.9, 20.0-29.9, and ≥ 30 U/mL to the reference (< 10 U/mL) were 1.08 (1.04-1.13), 1.18 (1.07-1.30), and 1.64 (1.35-1.99), respectively, among men. However, this association was not observed in women. The association between CA19-9 category and incident T2D significantly differed by sex (Pinteraction = 0.006). Among young and middle-aged Korean adults, elevated CA19-9 levels were significantly associated with increased risk of type 2 diabetes in men but not in women. Elevated CA19-9 levels in men could be a useful marker for identifying individuals at high risk of developing T2D. Evaluation approaches for individuals with elevated CA19-9 levels should be sex-specific.
Collapse
Affiliation(s)
- Sujeong Shin
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea.
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Samsung Main Building B2, 250, Taepyung-ro 2ga, Seoul, Jung-gu, 04514, South Korea.
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea.
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Samsung Main Building B2, 250, Taepyung-ro 2ga, Seoul, Jung-gu, 04514, South Korea.
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
39
|
Sun Y, Zhang L, Huang JQ, Su J, Cui LG. Non-invasive diagnosis of pancreatic steatosis with ultrasound images using deep learning network. Heliyon 2024; 10:e37580. [PMID: 39296003 PMCID: PMC11409133 DOI: 10.1016/j.heliyon.2024.e37580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Objective This study aimed to verify whether pancreatic steatosis (PS) is an independent risk factor for type 2 diabetes mellitus (T2DM). We also developed and validated a deep learning model for the diagnosis of PS using ultrasonography (US) images based on histological classifications. Methods In this retrospective study, we analysed data from 139 patients who underwent US imaging of the pancreas followed by pancreatic resection at our medical institution. Logistic regression analysis was employed to ascertain the independent predictors of T2DM. The diagnostic efficacy of the deep learning model for PS was assessed using receiver operating characteristic curve analysis and compared with traditional visual assessment methodology in US imaging. Results The incidence rate of PS in the study cohort was 64.7 %. Logistic regression analysis revealed that age (P = 0.003) and the presence of PS (P = 0.048) were independent factors associated with T2DM. The deep learning model demonstrated robust diagnostic capabilities for PS, with areas under the curve of 0.901 and 0.837, sensitivities of 0.895 and 0.920, specificities of 0.700 and 0.765, accuracies of 0.814 and 0.857, and F1-scores of 0.850 and 0.885 for the training and validation cohorts, respectively. These metrics significantly outperformed those of conventional US imaging (P < 0.001 and P = 0.045, respectively). Conclusion The deep learning model significantly enhanced the diagnostic accuracy of conventional ultrasound for PS detection. Its high sensitivity could facilitate widespread screening for PS in large populations, aiding in the early identification of individuals at an elevated risk for T2DM in routine clinical practice.
Collapse
Affiliation(s)
- Yang Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Li Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Jian-Qiu Huang
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Jing Su
- Department of Pathology, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Li-Gang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| |
Collapse
|
40
|
Lahooti A, Rizvi A, Canakis A, Akagbosu C, Johnson KE, Hassan K, Lahooti I, Abu-Hammour M, Dawod E, Dawod Q, Newberry C, Sampath K, Carr-Locke D, Mahadev S, Afaneh C, Dakin G, Kumar S, Yeung M, Barenbaum S, Tchang B, Shukla AP, Aronne LJ, Sharaiha RZ. Navigating the Predictive Landscape: DiaRem's Role in Unveiling Outcomes for Diabetes Remission following ESG. Obes Surg 2024; 34:3358-3365. [PMID: 39117857 DOI: 10.1007/s11695-024-07408-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE Rising obesity and type 2 diabetes mellitus (T2DM) rates can be mitigated by various strategies, with a 10% total body weight loss (TBWL) threshold often required for T2DM remission. T2DM remission rates after bariatric surgery like Roux-en-Y gastric bypass (RYGB) are well established; endoscopic sleeve gastroplasty (ESG) is a less invasive option that averages 15% TBWL and allows for T2DM remission. This study explores the DiaRem (Diabetes Remission post-RYGB) score's ability to predict T2DM remission 1-year post-ESG. MATERIALS AND METHODS We conducted a retrospective cohort study on 39 individuals with T2DM who underwent ESG. Age, utilization of diabetes medications, insulin administration, and hemoglobin A1c levels were used to calculate the DiaRem score. The area under the receiver operating characteristic curve (AUC) was employed to evaluate the discriminative ability of DiaRem in distinguishing diabetes remission. RESULTS Among the 39 patients with a median hemoglobin A1c of 6.7, 12.8% required insulin, and 43.6% used diabetes medication. At 1-year post-ESG, 69.2% of patients experienced diabetes remission with a median %TWBL of 12.7. The DiaRem score's ability to detect diabetes resolution for ESG patients had a sensitivity of 100% and a specificity of 58.3%, at the optimal cutoff value of 10. The AUC was 0.779 (95% CI 0.546-0.959). CONCLUSION Our study demonstrated the DiaRem score's predictive value for T2DM remission post-ESG, highlighting its utility in clinical decision-making for ESG-related outcomes. Further investigation is needed to identify alternative indicators that may enhance predictive accuracy, thus refining personalized decision-making for this patient group.
Collapse
Affiliation(s)
- Ali Lahooti
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Anam Rizvi
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Andrew Canakis
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cynthia Akagbosu
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Kate E Johnson
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Kamal Hassan
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Ila Lahooti
- Ohio University Heritage College of Osteopathic Medicine, Dublin, OH, USA
| | - Mohamed Abu-Hammour
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Enad Dawod
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Qais Dawod
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Carolyn Newberry
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Kartik Sampath
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - David Carr-Locke
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - SriHari Mahadev
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | | | - Gregory Dakin
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Sonal Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA
| | - Michele Yeung
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sarah Barenbaum
- Division of Endocrinology, Diabetes and Metabolism, Comprehensive Weight Control Center, Weill Cornell Medicine, New York, NY, USA
| | - Beverly Tchang
- Division of Endocrinology, Diabetes and Metabolism, Comprehensive Weight Control Center, Weill Cornell Medicine, New York, NY, USA
| | - Alpana P Shukla
- Division of Endocrinology, Diabetes and Metabolism, Comprehensive Weight Control Center, Weill Cornell Medicine, New York, NY, USA
| | - Louis J Aronne
- Division of Endocrinology, Diabetes and Metabolism, Comprehensive Weight Control Center, Weill Cornell Medicine, New York, NY, USA
| | - Reem Z Sharaiha
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, 4th Floor, New York, NY, 10021, USA.
| |
Collapse
|
41
|
Yuan W, Ran Y, Wang J, Pei F, Cui L, Chen S, Wu S, Zhou L. Mediating effect of diabetes on the relationship between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease: a prospective cohort study. Eur J Gastroenterol Hepatol 2024; 36:1133-1140. [PMID: 39101442 DOI: 10.1097/meg.0000000000002794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
OBJECTIVE This study explored the mediating effect of diabetes on the relationship between nonalcoholic fatty liver disease (NAFLD) and atherosclerotic cardiovascular disease (ASCVD). METHODS In this prospective community cohort study, 82 975 participants were enrolled, with the primary outcome being the incidence of new-onset ASCVD. Using the Cox proportional hazards model, the hazard ratio (HR) and 95% confidence interval (CI) for ASCVD occurrence were computed between NAFLD and non-NAFLD groups. The correlation between NAFLD and diabetes was assessed using a binary logistic regression model, and that between NAFLD, diabetes and ASCVD using a mediation model. RESULTS During follow-up, 9471 ASCVD cases were observed. Compared with individuals without NAFLD, those with NAFLD showed an increased ASCVD risk (HR: 1.424; 95% CI: 1.363-1.488; P < 0.001). Stratifying NAFLD based on metabolic subphenotypes revealed a higher ASCVD risk in the NAFLD combined with diabetes subgroup than in the non-NAFLD subgroup (HR: 1.960; 95% CI: 1.817-2.115; P < 0.001). NAFLD was positively associated with baseline diabetes (odds ratio: 2.983; 95% CI: 2.813-3.163; P < 0.001). Furthermore, NAFLD severity was positively correlated with diabetes risk. Mediation analysis indicated that diabetes partially mediated the effect of NAFLD on ASCVD incidence, accounting for 20.33% of the total effect. CONCLUSION NAFLD is an independent predictor of increased ASCVD risk, which may be slightly mediated by diabetes in patients with NAFLD. Evaluating NAFLD and diabetes may be crucial in the early screening and prevention of ASCVD.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
- Rheumatology and Immunology
| | - Ying Ran
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
| | | | | | | | - Shuohua Chen
- Cardiology, Kailuan General Hospital, Tangshan, China
| | - Shouling Wu
- Cardiology, Kailuan General Hospital, Tangshan, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
| |
Collapse
|
42
|
Shamanna P, Joshi S, Dharmalingam M, Vadavi A, Keshavamurthy A, Shah L, Samajdar SS, Mechanick JI. Digital Twin in Managing Hypertension Among People With Type 2 Diabetes: 1-Year Randomized Controlled Trial. JACC. ADVANCES 2024; 3:101172. [PMID: 39372467 PMCID: PMC11450914 DOI: 10.1016/j.jacadv.2024.101172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 10/08/2024]
Abstract
Background Digital twin (DT)-guided lifestyle changes induce type 2 diabetes (T2D) remission but effects on hypertension (HTN) in this population are unknown. Objectives The purpose of this study was to assess effects of DT vs standard of care (SC) on blood pressure (BP), anti-HTN medication, HTN remission, and microalbuminuria in participants with T2D. Methods This is a secondary analysis of a randomized controlled trial in India of 319 participants with T2D. Participants were randomized to DT group (N = 233), which used artificial intelligence-enabled DT technology, or SC group (N = 86). A Home Blood Pressure Monitoring system guided anti-HTN medication adjustments. BP, anti-HTN medications, HTN remission rates, and microalbuminuria were compared between groups. Results Among the 319 participants, 44 in DT and 15 in SC group were on anti-HTN medications, totaling 59 (18.4%) participants. DT group achieved significant reductions in systolic blood pressure (-7.6 vs -3.2 mm Hg; P < 0.007) and diastolic blood pressure (-4.3 vs -2.2 mm Hg; P = 0.046) after 1 year compared with SC group. 68.2% of DT group remained off anti-HTN medications compared to none in SC group. Among participants with HTN, DT subgroup achieved higher rates of normotension (40.9% vs 6.7%; P = 0.0009) and HTN remission (50% vs 0%; P < 0.0001) than SC subgroup. DT group had a higher rate of achieving normoalbuminuria (92.4% vs 83.1%; P = 0.018) at 1 year compared with SC group. Conclusions Artificial intelligence -enabled DT technology is more effective than SC in reducing BP and anti-HTN medications and inducing HTN remission and normoalbuminuria in participants with HTN and T2D. (A Novel WholeBody Digital Twin Enabled Precision Treatment for Reversing Diabetes; CTRI/2020/08/027072).
Collapse
Affiliation(s)
- Paramesh Shamanna
- Department of Diabetes, Bangalore Diabetes Centre, Bangalore, Karnataka, India
| | - Shashank Joshi
- Department of Diabetology and Endocrinology, Lilavati Hospital and Research Center, Mumbai, India
| | - Mala Dharmalingam
- MS Ramaiah Medical College, Bangalore Endocrinology & Diabetes Research Centre, Bangalore, Karnataka, India
| | - Arun Vadavi
- Department of Diabetes, Sudha Prevention Centre, Bangalore, Karnataka, India
| | | | - Lisa Shah
- Twin Health, Mountain View, California
| | - Shambo Samrat Samajdar
- Department of Clinical and Experimental Pharmacology, Calcutta School of Tropical Medicine, Kolkata, India
| | - Jeffrey I. Mechanick
- The Marie-Josee and Henry R. Kravis Center for Cardiovascular Health at Mount Sinai Fuster Heart Hospital, New York City, New York, USA
| |
Collapse
|
43
|
Strati M, Moustaki M, Psaltopoulou T, Vryonidou A, Paschou SA. Early onset type 2 diabetes mellitus: an update. Endocrine 2024; 85:965-978. [PMID: 38472622 DOI: 10.1007/s12020-024-03772-w] [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: 11/16/2023] [Accepted: 03/02/2024] [Indexed: 03/14/2024]
Abstract
The incidence and prevalence of type 2 diabetes mellitus (T2DM) in young individuals (aged <40 years) have significantly increased in recent years, approximating two to threefold increase in the respective rates. Numerous risk factors including severe obesity, family history, ethnicity, maternal diabetes or gestational diabetes, and female sex contribute to a younger age of onset. In terms of pathogenesis, impaired insulin secretion is the key operating mechanism, alongside with ectopic adiposity-related insulin resistance. T2DM diagnosis in a young adult requires the exclusion of type 1 diabetes mellitus (T1DM), latent autoimmune diabetes of adults (LADA) and maturity-onset diabetes of the young (MODY). The establishment of such diagnosis is critical for prognosis, because early-onset T2DM is associated with rapid deterioration in pancreatic β-cell secretory function leading to earlier initiation of insulin therapy. Furthermore, mortality and lifetime risk of developing complications, especially microvascular, is increased in these patients compared to both later-onset T2DM and T1DM patients; also, the latter are often developed earlier in the course of disease. The management of early-onset T2DM follows the same guidelines as in later-onset T2DM; yet patients aged 18-39 years are underrepresented in the big clinical trials on which the development of guidelines is based. Finally, young people with T2DM face significant challenges associated with social determinants, which compromise their adherence to therapy and induce diabetes distress. Future research focusing on the pathogenesis of β-cell decline and complications, as well as on specific treatment shall lead to better understanding and management of early-onset T2DM.
Collapse
Affiliation(s)
- Myrsini Strati
- School of Medicine, University of Patras, Patras, Greece
| | - Melpomeni Moustaki
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
44
|
Dash S. Opportunities to optimize lifestyle interventions in combination with glucagon-like peptide-1-based therapy. Diabetes Obes Metab 2024; 26 Suppl 4:3-15. [PMID: 39157881 DOI: 10.1111/dom.15829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024]
Abstract
Obesity is a chronic multi-system disease and major driver of type 2 diabetes and cardiometabolic disease. Nutritional interventions form the cornerstone of obesity and type 2 diabetes management. Some interventions such as Mediterranean diet can reduce incident cardiovascular disease, probably independently of weight loss. Weight loss of 5% or greater can improve many adiposity-related comorbidities. Although this can be achieved with lifestyle intervention, it is often difficult to sustain in the longer term due to adaptive endocrine changes. In recent years glucagon-like-peptide-1 receptor agonists (GLP-1RAs) have emerged as effective treatments for both type 2 diabetes and obesity. Newer GLP-1RAs can achieve average weight loss of 15% or greater and improve cardiometabolic health. There is heterogeneity in the weight loss response to GLP-1RAs, with a substantial number of patients unable to achieve 5% or greater weight. Weight loss, on average, is lower in older adults, male patients and people with type 2 diabetes. Mechanistic studies are needed to understand the aetiology of this variable response. Gastrointestinal side effects leading to medication discontinuation are a concern with GLP-1RA treatment, based on real-world data. With weight loss of 20% or higher with newer GLP-1RAs, nutritional deficiency and sarcopenia are also potential concerns. Lifestyle interventions that may potentially mitigate the side effects of GLP-1RA treatment and enhance weight loss are discussed here. The efficacy of such interventions awaits confirmation with well-designed randomized controlled trials.
Collapse
Affiliation(s)
- Satya Dash
- Division of Endocrinology, University Health Network & University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Taylor R. Understanding the cause of type 2 diabetes. Lancet Diabetes Endocrinol 2024; 12:664-673. [PMID: 39038473 DOI: 10.1016/s2213-8587(24)00157-8] [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: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024]
Abstract
Type 2 diabetes has long been thought to have heterogenous causes, even though epidemiological studies uniformly show a tight relationship with overnutrition. The twin cycle hypothesis postulated that interaction of self-reinforcing cycles of fat accumulation inside the liver and pancreas, driven by modest but chronic positive calorie balance, could explain the development of type 2 diabetes. This hypothesis predicted that substantial weight loss would bring about a return to the non-diabetic state, permitting observation of the pathophysiology determining the transition. These changes were postulated to reflect the basic mechanisms of causation in reverse. A series of studies over the past 15 years has elucidated these underlying mechanisms. Together with other research, the interaction of environmental and genetic factors has been clarified. This knowledge has led to successful implementation of a national programme for remission of type 2 diabetes. This Review discusses the paucity of evidence for heterogeneity in causes of type 2 diabetes and summarises the in vivo pathophysiological changes, which cause this disease of overnutrition. Type 2 diabetes has a homogenous cause expressed in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Newcastle Magnetic Resonance Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
46
|
Boocock RC, Haste A, Moore HJ, Lake AA. Barriers and enablers to engagement with a type 2 diabetes remission project in the North East of England: qualitative perspectives of patients. J Nutr Sci 2024; 13:e28. [PMID: 39776522 PMCID: PMC11704941 DOI: 10.1017/jns.2024.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/20/2024] [Accepted: 03/29/2024] [Indexed: 01/11/2025] Open
Abstract
This qualitative research sought to identify factors influencing patient choice of, and patient-related internal and external enablers and barriers to engagement with, type 2 diabetes (T2D) remission strategies offered by the Remission in diabetes (REMI.D) project. Patients had a choice of three diets: Total Diet Replacement (TDR)-Formula Food Products, TDR-Food, and Healthy lifestyle approach; and three activity pathways: Everyday life, General Practitioner referral, and Social hub. Semi-structured interviews were recorded and transcribed. Thematic analysis used the Framework Method and NVivo 12 to assist with generation and organisation of codes, inductive and deductive (Theoretical Domains Framework). The REMI.D project was a place-based approach (place in this case being defined as two local authorities with significant rates of deprivation) situated in the North East of England. Twenty patients out of a possible 65 patients took part. Areas of interest included: patient choice, patient intention, patient adherence, patient non-adherence, and patient stigma. Addition of a more moderate dietary strategy (not dissimilar to the diet in the Healthier You NHS Diabetes Prevention Programme) to the existing NHS England T2D Path to Remission programme may enable more patients to achieve remission or delayed progression with deprescribing of diabetes medications. Embedding a tailored physical activity path within or as a bolt-on to the NHS programme requires consideration. Limited resources should be targeted towards patients who identify with more barriers or fewer opportunities for health behaviour modification. Further research on use of virtual programmes in deprived areas is warranted.
Collapse
Key Words
- COM-B:, Capability Opportunity Motivation – Behaviour
- COVID-19:, Coronavirus
- DIADEM, Diabetes Intervention Accentuating Diet and Enhancing Metabolism
- DIAMOND, Diabetes Education and Self-Management for Ongoing and Newly Diagnosed
- DiRECT, Diabetes Remission Clinical Trial
- GP:, General practitioner
- IMD:, Index of multiple deprivation
- NIHR:, National Institute for Health Research
- Patient interviews
- Qualitative research
- REMI.D:, Remission in diabetes
- Remission strategy
- T2D:, Type 2 diabetes
- TDF:, Theoretical domains framework
- TDR:, Total diet replacement
- Type 2 diabetes
Collapse
Affiliation(s)
- Ruth C. Boocock
- School of Health & Life Sciences, Teesside University, Middlesbrough, UK
- Fuse, Centre for Translational Research in Public Health, University of Newcastle, Newcastle upon Tyne, UK
| | - Anna Haste
- Fuse, Centre for Translational Research in Public Health, University of Newcastle, Newcastle upon Tyne, UK
- School of Social Sciences, Humanities & Law, Teesside University, Middlesbrough, UK
| | - Helen J. Moore
- Fuse, Centre for Translational Research in Public Health, University of Newcastle, Newcastle upon Tyne, UK
- School of Social Sciences, Humanities & Law, Teesside University, Middlesbrough, UK
| | - Amelia A. Lake
- School of Health & Life Sciences, Teesside University, Middlesbrough, UK
- Fuse, Centre for Translational Research in Public Health, University of Newcastle, Newcastle upon Tyne, UK
| |
Collapse
|
47
|
Diamond C, Pansini M, Hamid A, Eichert N, Pandya P, Ali SN, Kemp GJ, Thanabalasingham G, Thomaides Brears H, Cuthbertson DJ. Quantitative Imaging Reveals Steatosis and Fibroinflammation in Multiple Organs in People With Type 2 Diabetes: A Real-World Study. Diabetes 2024; 73:1285-1299. [PMID: 38748492 PMCID: PMC11262045 DOI: 10.2337/db23-0926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/05/2024] [Indexed: 07/21/2024]
Abstract
We aimed to determine the extent of multiorgan fat accumulation and fibroinflammation in individuals living with type 2 diabetes. We deeply phenotyped individuals with type 2 diabetes (134 from secondary care, 69 from primary care) with multiorgan, quantitative, multiparametric MRI and compared with 134 matched control individuals without diabetes and 92 control individuals with normal weight. We examined the impact of diabetes duration, obesity status, and glycemic control. Ninety-three of the individuals with type 2 diabetes were reevaluated at 7 months (median). Multiorgan abnormalities were more common in individuals with type 2 diabetes (94%) than in age- and BMI-matched healthy individuals or healthy individuals with normal weight. We demonstrated a high burden of combined steatosis and fibroinflammation within the liver, pancreas, and kidneys (41%, 17%, and 10%) associated with visceral adiposity (73%) and poor vascular health (82%). Obesity was most closely associated with advanced liver disease, renal and visceral steatosis, and multiorgan abnormalities, while poor glycemic control was associated with pancreatic fibroinflammation. Pharmacological therapies with proven cardiorenal protection improved liver and vascular health unlike conventional glucose-lowering treatments, while weight loss or improved glycemic control reduced multiorgan adiposity (P ≤ 0.01). Quantitative imaging in people with type 2 diabetes highlights widespread organ abnormalities and may provide useful risk and treatment stratification. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | - Michele Pansini
- Perspectum, Ltd., Oxford, U.K
- Clinica Di Radiologia EOC, Istituto Di Imaging Della Svizzera Italiana, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Azlinda Hamid
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, U.K
| | - Nicole Eichert
- Perspectum, Ltd., Oxford, U.K
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, U.K
| | | | - Sarah N. Ali
- Royal Free London NHS Foundation Trust, London, U.K
| | - Graham J. Kemp
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, U.K
| | - Gaya Thanabalasingham
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Oxford University Hospitals NHS Foundation Trust, Oxford, U.K
| | | | - Daniel J. Cuthbertson
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, U.K
- University Hospital Aintree, Liverpool University Hospitals NHS Foundation Trust, Liverpool, U.K
| |
Collapse
|
48
|
Huttasch M, Roden M, Kahl S. Obesity and MASLD: Is weight loss the (only) key to treat metabolic liver disease? Metabolism 2024; 157:155937. [PMID: 38782182 DOI: 10.1016/j.metabol.2024.155937] [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: 08/29/2023] [Revised: 04/25/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) closely associates with obesity and type 2 diabetes. Lifestyle intervention and bariatric surgery aiming at substantial weight loss are cornerstones of MASLD treatment by improving histological outcomes and reducing risks of comorbidities. Originally developed as antihyperglycemic drugs, incretin (co-)agonists and SGLT2 inhibitors also reduce steatosis and cardiorenovascular events. Certain incretin agonists effectively improve histological features of MASLD, but not fibrosis. Of note, beneficial effects on MASLD may not necessarily require weight loss. Despite moderate weight gain, one PPARγ agonist improved adipose tissue and MASLD with certain benefit on fibrosis in post-hoc analyses. Likewise, the first THRβ-agonist was recently provisionally approved because of significant improvements of MASLD and fibrosis. We here discuss liver-related and metabolic effects induced by different MASLD treatments and their association with weight loss. Therefore, we compare results from clinical trials on drugs acting via weight loss (incretin (co)agonists, SGLT2 inhibitors) with those exerting no weight loss (pioglitazone; resmetirom). Furthermore, other drugs in development directly targeting hepatic lipid metabolism (lipogenesis inhibitors, FGF21 analogs) are addressed. Although THRβ-agonism may effectively improve hepatic outcomes, MASLD treatment concepts should consider all cardiometabolic risk factors for effective reduction of morbidity and mortality in the affected people.
Collapse
Affiliation(s)
- Maximilian Huttasch
- 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), Partner Düsseldorf, München-Neuherberg, 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 (DZD), Partner Düsseldorf, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Sabine Kahl
- 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), Partner Düsseldorf, München-Neuherberg, Germany.
| |
Collapse
|
49
|
Wang R, Mijiti S, Xu Q, Liu Y, Deng C, Huang J, Yasheng A, Tian Y, Cao Y, Su Y. The Potential Mechanism of Remission in Type 2 Diabetes Mellitus After Vertical Sleeve Gastrectomy. Obes Surg 2024; 34:3071-3083. [PMID: 38951388 DOI: 10.1007/s11695-024-07378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
In recent years, there has been a gradual increase in the prevalence of obesity and type 2 diabetes mellitus (T2DM), with bariatric surgery remaining the most effective treatment strategy for these conditions. Vertical sleeve gastrectomy (VSG) has emerged as the most popular surgical procedure for bariatric/metabolic surgeries, effectively promoting weight loss and improving or curing T2DM. The alterations in the gastrointestinal tract following VSG may improve insulin secretion and resistance by increasing incretin secretion (especially GLP-1), modifying the gut microbiota composition, and through mechanisms dependent on weight loss. This review focuses on the potential mechanisms through which the enhanced action of incretin and metabolic changes in the digestive system after VSG may contribute to the remission of T2DM.
Collapse
Affiliation(s)
- Rongfei Wang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Salamu Mijiti
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Qilin Xu
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Yile Liu
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Chaolun Deng
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Jiangtao Huang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Abudoukeyimu Yasheng
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Yunping Tian
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China.
| | - Yanlong Cao
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China.
| | - Yonghui Su
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
50
|
Kurnikowski A, Werzowa J, Hödlmoser S, Krenn S, Paschen C, Mussnig S, Tura A, Harreiter J, Krebs M, Song PX, Eller K, Pascual J, Budde K, Hecking M, Schwaiger E. Continuous Insulin Therapy to Prevent Post-Transplant Diabetes Mellitus: A Randomized Controlled Trial. Kidney Med 2024; 6:100860. [PMID: 39157193 PMCID: PMC11326904 DOI: 10.1016/j.xkme.2024.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
Rationale & Objectives Hyperglycemia is frequently observed early after transplantation and associated with development of post-transplant diabetes mellitus (PTDM). Here, we assessed continuous subcutaneous insulin infusion (CSII) targeting afternoon hyperglycemia. Study Design Open-label randomized parallel 3-arm design. Settings & Participants In total, 85 kidney transplant recipients without previous diabetes diagnosis were randomized to postoperative CSII therapy, basal insulin, or control. Interventions Insulin was to be initiated at afternoon capillary blood glucose level of ≥140 mg/dL (7.8 mmol/L; CSII and basal insulin) or fasting plasma glucose level of ≥200 mg/dL (11.1 mmol/L; control). Outcomes Hemoglobin A1c (HbA1c) levels at 3 months post-transplant (primary endpoint). PTDM assessed using oral glucose tolerance test at 12 and 24 months. Results CSII therapy lasted until median day 18 and maximum day 88. The median HbA1c value at month 3 was 5.6% (38 mmol/mol) in the CSII group versus 5.7% (39 mmol/mol) in the control group (P = 0.70) and 5.4% (36 mmol/mol) in the basal insulin group (P = 0.02). At months 12 and 24, the odds for PTDM were similar compared with the control group (odds ratios [95% confidence intervals], 0.80 [0.18-3.49] and 0.71 [0.15-3.16], respectively) and the basal insulin group (0.96 [0.18-5.68] and 1.51 [0.24-12.84], respectively). Mild hypoglycemia events occurred in the CSII and the basal insulin groups. Limitations This study is limited by outdated insulin pump technology, frequent discontinuations of CSII, a complex protocol, and concerns regarding reliability of HbA1c measurements. Conclusions CSII therapy was not superior at reducing HbA1c levels at month 3 or PTDM prevalence at months 12 and 24 compared with the control or basal insulin group.
Collapse
Affiliation(s)
- Amelie Kurnikowski
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Johannes Werzowa
- Ludwig Boltzmann Institute of Osteology, Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, Vienna, Austria
- First Medical Department, Hanusch Hospital, Vienna, Austria
| | - Sebastian Hödlmoser
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Simon Krenn
- Center for Health & Bioresources, Medical Signal Analysis, Austrian Institute of Technology GmbH, Vienna, Austria
| | - Christopher Paschen
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sebastian Mussnig
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andrea Tura
- CNR Institute of Neuroscience, Padova, Italy
| | - Jürgen Harreiter
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Medicine, Landesklinikum Scheibbs, Scheibbs, Austria
| | - Michael Krebs
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Peter X.K. Song
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Kathrin Eller
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Julio Pascual
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Klemens Budde
- Medizinische Klinik m. S. Nephrologie, Charité Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Manfred Hecking
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
- Kuratorium for Dialysis and Kidney Transplantation (KfH) e.V., Germany
| | - Elisabeth Schwaiger
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I: Cardiology and Nephrology, Hospital of the Brothers of St. John of God, Eisenstadt, Austria
| |
Collapse
|