1
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
2
|
Agoncillo ML, Gao Z, De Kraker HC, McHardy SF, Messing RO, Small L, Schmitz-Peiffer C. Effects of a protein kinase C epsilon inhibitor on insulin signalling in lipid-treated HepG2 hepatocytes and glucose tolerance in fat-fed mice. Eur J Pharmacol 2025; 997:177465. [PMID: 40054721 DOI: 10.1016/j.ejphar.2025.177465] [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: 01/12/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
AIMS Protein kinase C epsilon (PKCε) plays a causative role in the development of glucose intolerance, and is a potential target for the treatment of type 2 diabetes. Here, we examined the effects of the PKCε inhibitor CIDD-0150612 (CP612) on insulin action in palmitate-treated HepG2 hepatocytes in vitro and on glucose homeostasis in fat-fed mice in vivo. METHODS HepG2 cells were treated with palmitate and CP612 and stimulated with insulin. Insulin signalling was examined by immunoblotting and glucose incorporation into glycogen was measured using glucose tracer. Mice were fed a high-fat diet and treated with CP612 prior to glucose tolerance tests and tissue harvest. Proteomic analysis of liver was carried out by mass spectrometry. RESULTS CP612 promoted Akt phosphorylation in a highly insulin-dependent manner and reversed the inhibition of insulin-stimulated Akt phosphorylation and glucose incorporation into glycogen by palmitate. Fat-fed mice treated with CP612 had reduced fat mass, but not lean mass, compared with vehicle-treated littermates. Mice treated acutely with CP612 exhibited elevated fasting blood glucose. However, mice studied 24h after the last dose had lower fasting glucose and improved glucose tolerance with a lower insulin excursion. Proteomic analysis of liver from CP612-treated fat-fed mice indicated a reduction in gluconeogenic gene expression and decreased phosphorylation of the transcription factor Foxk1. CONCLUSIONS The PKCε inhibitor CP612 had beneficial effects on insulin action in hepatocytes and on fat mass and glucose homeostasis in mice. Because certain effects were not previously observed in genetically PKCε-deficient mice, off-target effects may be partly responsible.
Collapse
Affiliation(s)
- Miguel L Agoncillo
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia.
| | - Zhongmin Gao
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia.
| | - Harmannus C De Kraker
- Department of Chemistry, Center for Innovative Drug Discovery, University of Texas San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| | - Stanton F McHardy
- Department of Chemistry, Center for Innovative Drug Discovery, University of Texas San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| | - Robert O Messing
- Department of Neuroscience, University of Texas at Austin, E 24th Street, Austin, TX, 78712, USA.
| | - Lewin Small
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia.
| | - Carsten Schmitz-Peiffer
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia; Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia; St Vincent's Clinical School, University of New South Wales, 390 Victoria St, Darlinghurst, NSW, NSW 2010, Australia.
| |
Collapse
|
3
|
Zhang X, Nguyen MH. Metabolic dysfunction-associated steatotic liver disease: A sexually dimorphic disease and breast and gynecological cancer. Metabolism 2025; 167:156190. [PMID: 40081614 DOI: 10.1016/j.metabol.2025.156190] [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: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become a global public health and economic burden worldwide in the past few decades. Epidemiological studies have shown that MASLD is a multisystem disease that is associated not only with liver-related complications but also with an increased risk of developing extrahepatic cancers. MASLD is a sexually dimorphic disease with sex hormones playing an important role in the development and progression of MASLD, especially by the levels and ratios of circulating estrogens and androgens. MASLD is associated with hormone-sensitive cancers including breast and gynecological cancer. The risk of breast and gynecological cancer is elevated in individuals with MASLD driven by shared metabolic risk factors including obesity and insulin resistance. Multiple potential mechanisms underline these associations including metabolic dysfunction, gut dysbiosis, chronic inflammation and dysregulated release of hepatokines. However, the effect of hormone therapy including hormone replacement therapy and anti-estrogen treatment on MASLD and female-specific cancers remains debatable at this time. This synopsis will review the associations between MASLD and breast and gynecological cancer, their underlying mechanisms, implications of hormonal therapies, and their future directions.
Collapse
Affiliation(s)
- Xinrong Zhang
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States; Department of Epidemiology and Population Health, Stanford University Medical Center, Palo Alto, CA, United States; Stanford Cancer Institute, Stanford University Medical Center, Palo Alto, CA, United States.
| |
Collapse
|
4
|
Liu Y, Yu Z, Wang X, Yuan MQ, Lu MJ, Gong MR, Li Q, Xia YB, Yang GH, Xu B, Litscher G, Xu TC. Neurophysiological mechanisms of electroacupuncture in regulating pancreatic function and adipose tissue expansion. World J Diabetes 2025; 16:101354. [DOI: 10.4239/wjd.v16.i5.101354] [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: 09/11/2024] [Revised: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Electroacupuncture (EA) has been recognized for its beneficial effects on glucolipid metabolism, potentially through the regulation of sensory nerve coordination. The expandability of peripancreatic adipose tissue (PAT) is implicated in the transition from obesity to type 2 diabetes mellitus (T2DM). However, the specific pancreatic responses to EA require further elucidation.
AIM To investigate the influence of EA on pancreatic glucolipid reduction level in a high-fat diet (HFD) rat model.
METHODS To delineate the precise pathway through which EA mediates interactions between PAT and islets, we assessed the expression levels of NGF, TRPV1, insulin, as well as other proteins in the pancreas and PAT. This approach enabled us to identify the acupoints that are most conducive to optimizing glycolipid metabolism.
RESULTS The ST25, LI11 and ST37 groups attenuated HFD-induced obesity and insulin resistance (IR) to distinct degrees, with ST25 group having the greatest effect. EA at ST25 was found to modify the local regulatory influence of PAT on the pancreatic intrinsic nervous system. Specifically, EA at ST25 obviously activated the TRPV1-CGRP-islet beta cell pathway, contributing to the relief of glucolipid metabolic stress. The beneficial effects were abrogated following the chemical silencing of TRPV1 sensory afferents, confirming their indispensable role in EA-mediated regulation of islet and PAT function. Furthermore, in TRPV1 knockout mice, a reduction in PAT inflammation was observed, along with the recovery of islet beta cell function. EA at LI11 and ST37 demonstrated anti-inflammatory properties and helped ameliorate IR.
CONCLUSION The PAT ecological niche influenced the progression from obesity to T2DM through various immunometabolic pathways. EA at ST25 could regulate glucolipid metabolism via the TRPV1-CGRP-islet beta cell pathway.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Xuan Wang
- College of Traditional Chinese Medicine, Jiangsu Vocational College of Medicine, Yancheng 224000, Jiangsu Province, China
| | - Ming-Qian Yuan
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Meng-Jiang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Mei-Rong Gong
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - You-Bing Xia
- Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Guan-Hu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH 45701, United States
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Gerhard Litscher
- High-Tech Acupuncture and Digital Chinese Medicine, Swiss University of Traditional Chinese Medicine, Bad Zurzach 5530, Switzerland
| | - Tian-Cheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
5
|
Pan M, Qin G, Liu J, Yang M, Li X, Wu Z, Mai K, Zhang W. Establishment and Characterization of Hepatocyte Line from Turbot (Scophthalmus maximus L.) and Its Application in the Study of Glucose Metabolism. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2025; 27:77. [PMID: 40272575 DOI: 10.1007/s10126-025-10448-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
The present study was to establish a hepatocyte line and investigate its role in glucose metabolism. A continuous cell line, THL (turbot hepatocyte line), was established from the liver tissue of turbot (Scophthalmus maximus L.). It has been successfully passaged more than 60 generations. The THL cells showed an epithelial-like morphology and the normal chromosome number was 44. Different methods were used to identify the hepatocytes. Periodic acid-Schiff (PAS) staining for THL cells was positive, and two key functional proteins of hepatocytes, cytokeratin- 18 (CK- 18) and albumin (ALB), were detected in THL cells. The results of CCK- 8 indicated that a medium containing 15 mM glucose showed optimal cell viability of THL. Conversely, elevating glucose concentrations beyond 50 mM markedly impaired THL cell viability. Western blot and qRT-PCR were employed to assess the gene and protein expression in cells treated with varying concentrations of glucose. The results of cells incubated with 0 mM, 15 mM, and 50 mM glucose concentrations showed that compared with the 0 mM glucose group, 15 mM glucose could increase the gene expression of glucokinase (gk) and decrease the gene expression of cytosolic phosphoenolpyruvate carboxykinase (cpepck), mitochondrial phosphoenolpyruvate (mpepck), glucose- 6-phosphatase 1 (g6pase1), forkhead box o1 (foxo1), and glucose-regulated protein 78 (grp78). Compared to 15 mM glucose treatment, the expression of gk in the 50 mM group was significantly decreased, but the expression of cpepck, mpepck, g6pase1, foxo1, and grp78 was significantly increased. Moreover, the protein expression of FoxO1 and GRP78 in 50 mM treatment group was significantly increased compared to that in the15 mM group. In the present study, it was found that excessive glucose level can activate the pathways of FoxO1-mediated gluconeogenesis and GRP78-mediated endoplasmic reticulum stress and reduce the glycolytic pathway, thus disrupting the glucose homeostasis in hepatocytes.
Collapse
Affiliation(s)
- Mingzhu Pan
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
- Jiangsu Key Laboratory for Exploration and Utilization of Marine Wetland Biological Resources, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Gaochan Qin
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Jiahuan Liu
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Mengxi Yang
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Xinxin Li
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Zhenhua Wu
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Kangsen Mai
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
6
|
Wang Y, Yu Z, Yu L, Li C. Triglyceride-glucose index and triglyceride-to-high-density lipoprotein cholesterol ratio in predicting severity of acute pancreatitis: a cross-sectional clinical study. BMC Gastroenterol 2025; 25:226. [PMID: 40197175 PMCID: PMC11974065 DOI: 10.1186/s12876-025-03793-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND The aim of this study is to investigate the correlation of triglyceride-glucose (TyG) index and triglyceride-to-high-density lipoprotein cholesterol (TG/HDL-C) ratio with acute pancreatitis (AP), and to compare the predictive value of the two indexes for severe AP (SAP). METHODS This study was a clinical cross-sectional study. Spearman's correlation, logistic regression analysis and receiver operating characteristic (ROC) curves were used to investigate the relationship between the TyG index and TG/HDL-C ratio with SAP. RESULTS Of the 311 enrolled AP patients, the mean age was 62.59 ± 9.03 years, and 131 (42.12%) were male. A total of 34 (10.93%) patients met the diagnostic criteria for SAP. The results of Spearman's correlation showed that TyG index (Spearman rho = 0.262; p < 0.001), TG/HDL-C ratio (Spearman rho = 0.206; p < 0.001) were associated with SAP. Logistic regression analysis showed that TyG index was independently and positively correlated with SAP [odds ratio (OR), 4.311; 95% confidence interval (CI), 1.222-15.208; p = 0.023]. However, this association was not further confirmed on TG/HDL-C ratio (OR, 2.530; 95% CI, 0.883-7.251; p = 0.084). According to the ROC curve analysis, the area under the curve (AUC) for TyG index was 0.712 (p < 0.001), and the AUC for TG/HDL-C ratio was 0.691 (p < 0.001). CONCLUSIONS TyG index and TG/HDL-C ratio have different diagnostic values in AP patients. And the TyG index may be a more useful auxiliary tool for predicting SAP.
Collapse
Affiliation(s)
- Yakun Wang
- Department of Intensive Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medicine University, 1630 Huanding Road, Shangcheng District, Hangzhou, 310044, Zhejiang, China
| | - Zhenfei Yu
- Department of Intensive Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medicine University, 1630 Huanding Road, Shangcheng District, Hangzhou, 310044, Zhejiang, China
| | - Limei Yu
- Department of Intensive Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medicine University, 1630 Huanding Road, Shangcheng District, Hangzhou, 310044, Zhejiang, China
| | - Chen Li
- Department of Intensive Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medicine University, 1630 Huanding Road, Shangcheng District, Hangzhou, 310044, Zhejiang, China.
| |
Collapse
|
7
|
Leite JSM, Vilas-Boas EA, Takahashi HK, Munhoz AC, Araújo LCC, Carvalho CR, Jr JD, Curi R, Carpinelli AR, Cruzat V. Liver lipid metabolism, oxidative stress, and inflammation in glutamine-supplemented ob/ob mice. J Nutr Biochem 2025; 138:109842. [PMID: 39824260 DOI: 10.1016/j.jnutbio.2025.109842] [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: 06/05/2024] [Revised: 12/21/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Glutamine availability may be reduced in chronic diseases, such as type 2 diabetes mellitus (T2DM)-induced by obesity. Herein, the antioxidant, anti-inflammatory and lipid metabolism effects of chronic oral glutamine supplementation in its free and dipeptide form were assessed in ob/ob mice. Adult male C57BL/6J ob/ob mice were supplemented with L-alanyl-L-glutamine (DIP) or free L-glutamine (GLN) in the drinking water for 40 days, whilst C57BL/6J Wild-type lean (WT) and control ob/ob mice (CTRL) received fresh water only. Plasma and tissue (skeletal muscle and liver) glutamine levels, and insulin resistance parameters (e.g., GTT, ITT, insulin) were determined. Oxidative stress (e.g., GSH system, Nrf2 translocation), inflammatory (e.g., NFkB translocation, TNF-α gene expression) and lipid metabolism parameters (e.g., plasma and liver triglyceride levels, SRBP-1, FAS, ACC, and ChRBP gene expression) were also analyzed. CTRL ob/ob mice showed lower glutamine levels in plasma and tissue, as well as increased insulin resistance and fat in the liver. Conversely, chronic DIP supplementation restored glutamine levels in plasma and tissues, improved glucose homeostasis and reduced plasma and liver lipid levels. Also, Nrf2 restoration, reduced NFkB translocation, and lower TNF-α gene expression was observed in the DIP group. Interestingly, chronic free GLN only increased muscle glutamine stores but reduced overall insulin resistance, and attenuated plasma and liver lipid metabolic biomarkers. The results presented herein indicate that restoration of body glutamine levels reduces oxidative stress and inflammation in obese and T2DM ob/ob mice. This effect attenuated hepatic lipid metabolic changes observed in obesity.
Collapse
Affiliation(s)
- Jaqueline Santos Moreira Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Eloisa Aparecida Vilas-Boas
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Hilton K Takahashi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Layanne C C Araújo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Carla Roberta Carvalho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Jose Donato Jr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, ICAFE, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil; Instituto Butantan, São Paulo, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Vinicius Cruzat
- Faculty of Health, Southern Cross University, Gold Coast, Queensland, Australia.
| |
Collapse
|
8
|
Datta D, Kundu R, Basu R, Chakrabarti P. Pathophysiological hallmarks in type 2 diabetes heterogeneity (review). Diabetol Int 2025; 16:201-222. [PMID: 40166449 PMCID: PMC11954762 DOI: 10.1007/s13340-024-00783-w] [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: 03/01/2024] [Accepted: 10/21/2024] [Indexed: 01/03/2025]
Abstract
The mechanistic complexity in type 2 diabetes (T2DM) is primarily responsible for the degrees of heterogeneity and development of complications. A complex mode of interactions between different pathophysiological events and diabetogenic environmental factors support for the genesis of diabetes heterogeneity both in phenotypic and clinical contexts. The currently used diabetes classification strategies suffer from several inconsistencies that cannot fully capture the inherent heterogeneity among the diabetes patients. To effectively address this pathobiological and heterogeneity-related issue in diabetes research, the current review proposes nine pathophysiological hallmarks of T2DM that aims to mechanistically explain complexities of diabetes associated pathophysiological events and their underlying features. These pathophysiological hallmarks are pancreatic beta cell dysfunction, insulin sensitivity, insulin resistance, obesity, aging, subclinical inflammation, metabolic dysregulation, prothrombotic state induction and hypertension. Detail knowledge of these pathophysiological hallmarks with their key molecular mediators, influencing factors, clinical biomarkers and clinical assessment methodologies will greatly support precision medicine approaches in diabetes including patient stratification, subtype diagnosis and treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s13340-024-00783-w.
Collapse
Affiliation(s)
- Dipamoy Datta
- Computer Education Training Program, NICS Computer, Kolkata, 700032 India
- Department of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4 Raja SC Mullick Road, Kolkata, 700032 India
| | - Raja Kundu
- Computer Education Training Program, NICS Computer, Kolkata, 700032 India
| | - Rajdeep Basu
- Department of Endocrinology, Nil Ratan Sarkar Medical College, Kolkata, 700014 India
| | - Partha Chakrabarti
- Department of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4 Raja SC Mullick Road, Kolkata, 700032 India
| |
Collapse
|
9
|
Ni M, Chen Y, Gu W, Zhang Y, Xu M, Gu Y, Chen Y, Zhu Y, Wang X, Luo Y, Xu Y, Lin X, Zeng YA, Liu R, Wang J. Association Between Circulating Gremlin 2 and β-Cell Function Among Participants With Prediabetes and Type 2 Diabetes. J Diabetes 2025; 17:e70090. [PMID: 40270326 PMCID: PMC12018890 DOI: 10.1111/1753-0407.70090] [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: 03/06/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
AIM Circulating Gremlin 2 (Grem2) has recently been linked to human obesity, but its role in type 2 diabetes (T2D) remains unclear. This study aims to explore the association of circulating Grem2 with β-cell function. METHODS A post hoc analysis was conducted using data from three clinical trials, in which all participants underwent the oral glucose tolerance test (OGTT). Circulating Grem2 levels were measured at 0, 1, and 2 h during the OGTT. In Trial 1, Grem2 levels were compared between participants with T2D (n = 59) and without T2D (n = 119). We further examined changes in Grem2 levels in response to oral antidiabetic drugs in participants with T2D in Trial 2 (n = 67) and calorie restriction in participants with prediabetes in Trial 3 (n = 231). The relationship between Grem2 levels and β-cell function was analyzed across all trials. RESULTS Fasting and 1-h Grem2 levels were lower in participants with T2D compared with those without T2D (728 ± 25 vs. 649 ± 31 pg/mL, p = 0.020; 631 ± 26 vs. 537 ± 31 pg/mL, p = 0.007). Fasting Grem2 levels were restored after antidiabetic treatment (550 ± 12 vs. 575 ± 12 pg/mL, p = 0.019), and 1-h Grem2 levels increased following calorie restriction (1118 ± 89 vs. 1144 ± 90 vs. 1253 ± 89 pg/mL, p for trend = 0.002). The 1-h Grem2 levels were positively associated with β-cell function assessed by the oral disposition index and HOMA-β. CONCLUSION Reduced circulating Grem2 levels are associated with impaired β-cell function in T2D, and could be restored through antidiabetic interventions. TRIAL REGISTRATION ClinicalTrials.gov: NCT01959984, NCT01758471, NCT03856762.
Collapse
Affiliation(s)
- Mengshan Ni
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Yanru Chen
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Weiqiong Gu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Yifei Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Min Xu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Yanyun Gu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Yufei Chen
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Yinmeng Zhu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Xiao Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Yaogan Luo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Yu Xu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
- Key Laboratory of Systems BiologyHangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of SciencesHangzhouChina
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell BiologyUniversity of Chinese Academy of SciencesShanghaiChina
| | - Ruixin Liu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| | - Jiqiu Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghaiChina
| |
Collapse
|
10
|
Liu W, Jin R, Ma F, Zhao P, Su Y, Wang J, Zhang Y, Wang R, Zhu J, Liu X. Effects of Dioscorea opposita polysaccharides on insulin resistance and gut microbiota in high-fat-diet induced type 2 diabetic rats. Int J Biol Macromol 2025; 304:141004. [PMID: 39952518 DOI: 10.1016/j.ijbiomac.2025.141004] [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: 03/26/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
This study investigates the effects Dioscorea opposita polysaccharides (DOP) on insulin resistance, lipid metabolism, oxidative stress, and intestine microbiota in high-fat-diet and streptozotocin induced type 2 diabetes (T2DM) rats. Low dose (DOP-L, 200 mg/kg BW), high dose (DOP-H, 400 mg/kg BW) and D. opposita powder (DO, 400 mg/kg BW) were oral-administrated to T2DM rats. After 6 weeks of treatment, supplementation of DOP-H and DO improved body weight and glucose/lipid metabolism-related indicators, including glucagon-like peptide 1, total cholesterol and high-density lipoprotein cholesterol. DOP-H and DO suppressed liver oxidative stress through increasing the level of superoxide dismutase, catalase, glutathione and reducing malondialdehyde. DOP attenuated the pathological change in liver, such as hepatic steatosis, and thus improved the liver function. Furthermore, the anti-diabetic effects of DOP was correlated with alterations of the gut microbiota, including an increase in Firmicutes and Bacteroidetes and a decrease in Actinobacteria and Proteobacteria, which promoted a healthier gut environment. Further analysis of short-chain fatty acids and metabolites provided evidences of DOP's regulatory effects on cecal contents in T2DM rats. Therefore, DOP-H present decent effects on T2DM, suggesting that DOP can ameliorate the insulin resistance and restore blood lipid level of T2DM rats with high-fat-diet by regulating intestinal microbiota.
Collapse
Affiliation(s)
- Wei Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China; Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rumeng Jin
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Fanyi Ma
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China.
| | - Peng Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yuting Su
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Jiani Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Yun Zhang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Ruijiao Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Jinhua Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Xiuhua Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China.
| |
Collapse
|
11
|
Wang J, Liao M, Tong Z, Yuan S, Hu Z, Chen Z, Zeng F, Zou R, Chen D, Chen G, Wang Z, Liu W. Treadmill Exercise Modulates the Leptin/LepR/GSK-3β Signalling Pathway to Improve Leptin Sensitivity and Alleviate Neuroinflammation in High-Fat Diet-Fed APP/PS1 Mice. Mol Neurobiol 2025:10.1007/s12035-025-04853-1. [PMID: 40131695 DOI: 10.1007/s12035-025-04853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Neuroinflammation plays a critical role in the development of Alzheimer's disease (AD) and is closely associated with obesity. In AD, the fat cell-secreted protein leptin crosses the blood-brain barrier and protects against nerve damage. However, obesity may induce leptin resistance, reduce leptin sensitivity, stimulate excessive glial cell activation, promote inflammatory factor production and exacerbate brain inflammation. Unfortunately, the mechanism of interaction among high-fat diets, obesity, neuroinflammation and neurodegenerative diseases remains unclear. We investigated the changes in neuroinflammation and leptin sensitivity in the brains of wild-type and high-fat-diet-fed APP/PS1 transgenic mice. We explored the effects of treadmill exercise for 12 weeks on the leptin/LepR/GSK-3β signalling pathway and memory. The body weights of the high-fat-diet-fed mice increased, and elevated levels of markers for leptin resistance, including suppressor of signalling 3 (SOCS3), protein tyrosine phosphatase 1B (PTP1B) and proinflammatory factors such as tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6), were observed. After 12 weeks of aerobic exercise, the leptin mRNA and protein levels increased, GSK-3β protein expression decreased and the mean fluorescence intensities of brain microglial (IBA-1) and neuron markers (NeuN) decreased, indicating that exercise may activate the leptin/LepR/GSK-3β signalling pathway, reducing glial cell activation and inflammation. Our study revealed that obesity induces and exacerbates the AD-related neuroinflammatory response. Aerobic exercise activates the leptin/LepR/GSK-3β pathway to relieve neuroinflammation and protect nerve cells, alleviating AD-associated memory loss. These promising outcomes could inform the development of nondrug-based aerobic exercise interventions for the treatment of AD and associated cognitive disorders.
Collapse
Affiliation(s)
- Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Meiqing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Shunling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zelin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Ruihan Zou
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Dandan Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Gan Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Physical Education College, Hunan Normal University, Yuelu District, No. 437, Lushan South Road, Changsha, 410081, China.
| | - Zhiyuan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Physical Education College, Hunan Normal University, Yuelu District, No. 437, Lushan South Road, Changsha, 410081, China.
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Physical Education College, Hunan Normal University, Yuelu District, No. 437, Lushan South Road, Changsha, 410081, China.
| |
Collapse
|
12
|
El-Sehrawy AAMA, Khachatryan LG, Kubaev A, Rekha MM, Rab SO, Kaur M, Singh M, Srivastava M, Kadhim AJ, Zwamel AH. Triglyceride-glucose index: a potent predictor of metabolic risk factors and eating behavior patterns among obese individuals. BMC Endocr Disord 2025; 25:71. [PMID: 40098017 PMCID: PMC11917143 DOI: 10.1186/s12902-025-01887-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The strong potential of triglyceride to glucose index (TyG) in prediction of metabolic abnormalities is well identified in numerous disease including diabetes, metabolic syndrome and kidney disorders. However, no study is available to assess its validity and association with metabolic phenotype among obese individuals. In the current study, we aimed to evaluate the TyG index, its validity and association with metabolic parameters among obese individuals. METHODS AND MATERIALS In the current cross-sectional study, 300 obese individuals were enrolled. Their demographic, anthropometric measurements were done and laboratory parameters including serum lipids, glycemic markers and insulin resistance were evaluated. Blood pressure was also measured with standard methods. The TyG index was calculated as the ln (fasting triglyceride level [mg/dL] × fasting glucose level [mg/dL]/2). Eating pattern was measured with three factor eating behavior questionnaire (TFEQ). Receiver operator characteristic curve was used to assess the TyG validity. RESULTS Subjects at the higher TyG tertile had higher waist o hip ratio (WHR) and eating disorder compared with lowest tertiles. Also, those at the highest tertiles had significantly higher total cholesterol (TC), triglyceride (TG), and fasting blood sugar (FBS), and lower high density lipoprotein cholesterol (HDL). According to the ROC curve analysis for various metabolic parameters, TyG demonstrated the highest area under curve (AUC) value of 0.838 compared with other metabolic parameters in identification of metabolic syndrome. CONCLUSION The current study provides valuable insights into the relationship between TyG index, metabolic parameters, and eating behaviors among obese individuals.
Collapse
Affiliation(s)
| | - Lusine G Khachatryan
- Department of Pediatric Diseases, N.F. Filatov Clinical Institute of Children's Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, 140100, Uzbekistan.
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Safia Obaidur Rab
- Central Labs, King Khalid University, AlQura'a, P.O. Box 960, AlQura'a, Abha, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Manmeet Singh
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges- Jhanjeri, Mohali, Punjab, 140307, India
| | - Manish Srivastava
- Department of Endocrinology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
13
|
Lei Y, Zhang R, Li Y, Pang H, Fu Q, Chen C, Liu F. Pueraria Radix and Its Major Constituents Against Metabolic Diseases: Pharmacological Mechanisms and Potential Applications. Phytother Res 2025. [PMID: 40099674 DOI: 10.1002/ptr.8464] [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: 09/18/2024] [Revised: 12/28/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025]
Abstract
Metabolic diseases (MD), a series of chronic disorders, severely decrease the quality of life for patients but also cause a heavy economic burden. The ancient Chinese herb Pueraria Radix (PR) plays an important role in curing MD. Up to now, the bioactive compounds found in PR demonstrate effective actions in treating various metabolic disorders. This paper systematically summarizes the recent research advances on the pharmacological activities of PR and its constituents, explains the underlying mechanisms of preventing and treating MD. Besides, phytochemicals, drug delivery systems, clinical application, and safety of PR have been researched, hoping to provide valuable information for the future application, development, and improvement of PR as well as MD treatment. The information about PR was collected from various sources including classic books about Chinese herbal medicine and scientific databases including Web of Science, PubMed, ScienceDirect, Springer, ACS, SCOPUS, CNKI, Google Scholar, X-MOL, and WANFANG using keywords given and terms like pharmacological and phytochemical details of this plant. The chemical constituents isolated and identified from PR, such as isoflavones including puerarin, formononetin, daidzin, daidzein, genistein, and so forth, polysaccharides, alkaloids, starch, and other components have been proved to have the effect of anti-diabetic, anti-obesity, anti-atherosclerotic, anti-osteoporotic, anti-hypertensive, anti-hyperlipidemia, and anti-nonalcoholic fatty liver disease (NAFLD) through PI3K/Akt, Nrf2/HO-1, LOX-1/ROS/Akt/eNOS, ERK1/2-Nrf2, GLP-1R, Caspase, MAPK, NF-κB, and other anti-inflammatory and anti-oxidant signaling pathways. Also, the active contents of PR have been designed as drug delivery systems to improve the therapeutic effects of MD. It provides a preclinical basis for the efficacy of PR as an effective therapeutic agent for the prevention and treatment of MD. Even so, further studies are still needed to enhance bioavailability and expand clinical application.
Collapse
Affiliation(s)
- Yicheng Lei
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Ruiyuan Zhang
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Yan Li
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Huiwen Pang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - Qiang Fu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, China
| | - Chen Chen
- School of Biomedical Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Fang Liu
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Bazzazzadehgan S, Shariat-Madar Z, Mahdi F. Distinct Roles of Common Genetic Variants and Their Contributions to Diabetes: MODY and Uncontrolled T2DM. Biomolecules 2025; 15:414. [PMID: 40149950 PMCID: PMC11940602 DOI: 10.3390/biom15030414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) encompasses a range of clinical manifestations, with uncontrolled diabetes leading to progressive or irreversible damage to various organs. Numerous genes associated with monogenic diabetes, exhibiting classical patterns of inheritance (autosomal dominant or recessive), have been identified. Additionally, genes involved in complex diabetes, which interact with environmental factors to trigger the disease, have also been discovered. These genetic findings have raised hopes that genetic testing could enhance diagnostics, disease surveillance, treatment selection, and family counseling. However, the accurate interpretation of genetic data remains a significant challenge, as variants may not always be definitively classified as either benign or pathogenic. Research to date, however, indicates that periodic reevaluation of genetic variants in diabetes has led to more consistent findings, with biases being steadily eliminated. This has improved the interpretation of variants across diverse ethnicities. Clinical studies suggest that genetic risk information may motivate patients to adopt behaviors that promote the prevention or management of T2DM. Given that the clinical features of certain monogenic diabetes types overlap with T2DM, and considering the significant role of genetic variants in diabetes, healthcare providers caring for prediabetic patients should consider genetic testing as part of the diagnostic process. This review summarizes current knowledge of the most common genetic variants associated with T2DM, explores novel therapeutic targets, and discusses recent advancements in the pharmaceutical management of uncontrolled T2DM.
Collapse
Affiliation(s)
- Shadi Bazzazzadehgan
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Zia Shariat-Madar
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| | - Fakhri Mahdi
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
15
|
Kwon Y, Yoon H, Ha J, Lee HS, Pahk K, Kwon H, Kim S, Park S. Changes in pancreatic levodopa uptake in patients with obesity and new-onset type 2 diabetes: an 18F-FDOPA PET-CT study. Front Endocrinol (Lausanne) 2025; 16:1460253. [PMID: 40099262 PMCID: PMC11911206 DOI: 10.3389/fendo.2025.1460253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Levodopa (L-3,4-dihydroxyphenylalanine)g, a dopamine precursor that circulates in the peripheral region, is involved in pancreatic glycemic control. Although previous animal studies have shown that peripheral levodopa is correlated with insulin secretion in pancreatic beta cells, the mechanism by which the pancreas uses levodopa differently in humans with obesity and type 2 diabetes remains unknown. Our study aimed to observe how the pancreas uptakes and utilizes levodopa differently under obese and diabetic conditions. Materials and method 18F-fluoro-L-dopa positron emission tomography-computed tomography (18F-FDOPA PET-CT) was used to visualize how the human body uses levodopa under obese and diabetic conditions and presented its clinical implications. 10 patients were divided into 3 groups: 1) Group A, normal weight without type 2 diabetes; 2) Group B, obese without type 2 diabetes; and 3) Group C, obese with new-onset type 2 diabetes. All patients' lifestyle modification was conducted prior to 18F-FDOPA PET-CT, and plasma samples were collected to confirm changes in amino acid metabolites. Results Pancreatic levodopa uptake increased in obese patients with insulin resistance, whereas it decreased in obese patients with new-onset type 2 diabetes [standardized uptake value (SUV) mean in participants with normal weight, 2.6 ± 0.7; SUVmean in patients with obesity, 3.6 ± 0.1; SUVmean in patients with obesity and new-onset type 2 diabetes, 2.6 ± 0.1, P = 0.02]. Conclusions This suggested that the alterations in the functional capacity of pancreatic beta cells to take up circulating levodopa are potentially linked to the insulin resistance and the pathogenesis of type 2 diabetes. The differences in the uptake values between the groups implied that pancreatic levodopa uptake could be an early indicator of type 2 diabetes.
Collapse
Affiliation(s)
- Yeongkeun Kwon
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
- Gut & Metabolism Laboratory, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Foregut Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hanseok Yoon
- Division of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jane Ha
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Hyeon-Seong Lee
- Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Kisoo Pahk
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyunwoo Kwon
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sungeun Kim
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sungsoo Park
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
- Gut & Metabolism Laboratory, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Foregut Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Guzman H, Hasan LZ, Reid TJ. Treatment of Type 2 Diabetes in Patients with Obesity: A Review. Endocrinol Metab Clin North Am 2025; 54:163-173. [PMID: 39919872 DOI: 10.1016/j.ecl.2024.10.004] [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] [Indexed: 02/09/2025]
Abstract
Type 2 diabetes and obesity have some overlapping pathophysiology. This has allowed for the creation of therapies which are highly effective in treating both conditions. Weekly subcutaneous incretin agonists are preferred agents as they provide significant improvement in glycemic parameters, weight, and other comorbidities, like heart failure and reduce major adverse cardiovascular event. Bariatric surgery continues to show the most durable benefits for patients with both type 2 diabetes mellitus and obesity and should be considered in patients who are unable to meet goals with pharmacotherapy and lifestyle.
Collapse
Affiliation(s)
- Heidi Guzman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Columbia University College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Leen Z Hasan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Columbia University College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Tirissa J Reid
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Columbia University College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
17
|
Mather KJ, Mari A, Weerakkody G, Heise T, DeVries JH, Urva S, Coskun T, Milicevic Z, Haupt A, Thomas MK. Greater improvement in insulin sensitivity per unit weight loss associated with tirzepatide versus semaglutide: An exploratory analysis. Diabetes Obes Metab 2025; 27:1507-1514. [PMID: 39762971 DOI: 10.1111/dom.16159] [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: 08/26/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 02/08/2025]
Abstract
AIMS To explore the relationship between weight loss and insulin sensitivity in response to tirzepatide or semaglutide. MATERIALS AND METHODS We conducted a post hoc exploratory analysis of a 28-week, double-blind, randomized trial in people with type 2 diabetes treated with metformin, randomized to tirzepatide 15 mg, semaglutide 1 mg or placebo. We evaluated the relationship between change in body weight and change in insulin sensitivity determined from hyperinsulinemic euglycemic clamp (M value), or from mixed-meal tolerance testing (Matsuda index). RESULTS Tirzepatide was associated with a greater improvement than semaglutide in insulin sensitivity assessed using hyperinsulinemic euglycemic clamps (p < 0.001). With tirzepatide, improvements in insulin sensitivity were associated with percent change in weight (R = -0.656, p < 0.0001). With semaglutide, change in insulin sensitivity was less strongly correlated with percent change in weight (R = -0.268, p = 0.0820; p = 0.0242 vs. tirzepatide). In regression analyses, the slope of the relationship between change in M value and change in weight was statistically different between semaglutide and tirzepatide (p = 0.0461). These relationships were also assessed using the Matsuda index as the metric of insulin sensitivity, and using change in fat mass as the determinant of change in insulin sensitivity. CONCLUSIONS Improvement in insulin sensitivity was proportional to weight and fat loss, with greater strength of association with tirzepatide. In regression analysis, tirzepatide was associated with greater improvement in insulin sensitivity per unit weight loss than semaglutide. The greater improvement in insulin sensitivity following treatment with tirzepatide was not simply attributable to greater weight or fat loss.
Collapse
Affiliation(s)
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | | | | | - Shweta Urva
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Tamer Coskun
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Axel Haupt
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | |
Collapse
|
18
|
Wu Z, Liu Y, Wang B. The relationship between weight-adjusted waist index and peripheral artery disease. Front Nutr 2025; 12:1504896. [PMID: 40013161 PMCID: PMC11860073 DOI: 10.3389/fnut.2025.1504896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Background Obesity is a significant risk factor for peripheral arterial disease (PAD). The weight-adjusted waist index (WWI) is a novel obesity metric that better reflects abdominal obesity than traditional body mass index (BMI). However, research on the relationship between WWI and PAD remains scarce. Methods Relevant data from the NHANES 1999-2004 were selected. Multiple logistic regression and restricted cubic spline (RCS) analyses were used to assess the relationship between WWI and the risk of PAD. Additionally, the area under the curve (AUC) of the receiver operating characteristic (ROC) was used to evaluate the predictive ability of WWI for PAD. Results A total of 5,686 participants were included in the study, of whom 476 had PAD and 5,210 did not. The results of multiple logistic regression showed that WWI was significantly positively associated with the risk of PAD after full adjustment for covariates (OR: 1.407, 95% CI: 1.100-1.799). Additionally, compared to the first quartile of WWI, the risk of PAD significantly increased in the second quartile (OR: 2.042, 95% CI: 1.333-3.129), third quartile (OR: 2.134, 95% CI: 1.354-3.364), and fourth quartile (OR: 2.491, 95% CI: 1.435-4.325). The ROC results showed that the AUC value for WWI was 0.697, while the AUC value for BMI was 0.520. Compared to BMI, WWI has a better predictive value for the risk of PAD. Conclusion There is a significant positive correlation between WWI and the risk of PAD. For individuals with high WWI, efforts should be made to reduce WWI to prevent the onset of PAD.
Collapse
Affiliation(s)
- Zhe Wu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Liu
- Department of General Surgery, Vascular Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Bin Wang
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
19
|
Yoshiji S, Lu T, Butler-Laporte G, Carrasco-Zanini-Sanchez J, Su CY, Chen Y, Liang K, Willett JDS, Wang S, Adra D, Ilboudo Y, Sasako T, Koyama S, Nakao T, Forgetta V, Farjoun Y, Zeberg H, Zhou S, Marks-Hultström M, Machiela MJ, Kaalia R, Dashti H, Claussnitzer M, Flannick J, Wareham NJ, Mooser V, Timpson NJ, Langenberg C, Richards JB. Integrative proteogenomic analysis identifies COL6A3-derived endotrophin as a mediator of the effect of obesity on coronary artery disease. Nat Genet 2025; 57:345-357. [PMID: 39856218 PMCID: PMC11821532 DOI: 10.1038/s41588-024-02052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
Obesity strongly increases the risk of cardiometabolic diseases, yet the underlying mediators of this relationship are not fully understood. Given that obesity strongly influences circulating protein levels, we investigated proteins mediating the effects of obesity on coronary artery disease, stroke and type 2 diabetes. By integrating two-step proteome-wide Mendelian randomization, colocalization, epigenomics and single-cell RNA sequencing, we identified five mediators and prioritized collagen type VI α3 (COL6A3). COL6A3 levels were strongly increased by body mass index and increased coronary artery disease risk. Notably, the carboxyl terminus product of COL6A3, endotrophin, drove this effect. COL6A3 was highly expressed in disease-relevant cell types and tissues. Finally, we found that body fat reduction could reduce plasma levels of COL6A3-derived endotrophin, indicating a tractable way to modify endotrophin levels. In summary, we provide actionable insights into how circulating proteins mediate the effects of obesity on cardiometabolic diseases and prioritize endotrophin as a potential therapeutic target.
Collapse
Grants
- 169303 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- 365825 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- K99 HL169733 NHLBI NIH HHS
- 100558 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- 409511 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- 202460267 MEXT | Japan Society for the Promotion of Science (JSPS)
- Wellcome Trust
- The Richards research group is supported by the Canadian Institutes of Health Research (CIHR: 365825, 409511, 100558, 169303), the McGill Interdisciplinary Initiative in Infection and Immunity (MI4), the Lady Davis Institute of the Jewish General Hospital, the Jewish General Hospital Foundation, the Canadian Foundation for Innovation, the NIH Foundation, Cancer Research UK, Genome Québec, the Public Health Agency of Canada, McGill University, Cancer Research UK [grant number C18281/A29019] and the Fonds de Recherche Québec Santé (FRQS). J.B.R. is supported by an FRQS Mérite Clinical Research Scholarship. Support from Calcul Québec and Compute Canada is acknowledged. TwinsUK is funded by the Welcome Trust, Medical Research Council, European Union, the National Institute for Health Research (NIHR)-funded BioResource, Clinical Research Facility and Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust in partnership with King’s College London. NJT is a Wellcome Trust Investigator (202802/Z/16/Z), is the PI of the Avon Longitudinal Study of Parents and Children (MRC & WT 217065/Z/19/Z), is supported by the University of Bristol NIHR Biomedical Research Centre (BRC-1215-2001), the MRC Integrative Epidemiology Unit (MC_UU_00011/1) and works within the CRUK Integrative Cancer Epidemiology Programme (C18281/A29019).
- T.L. is supported by a Schmidt AI in Science Postdoctoral Fellowship, a Vanier Canada Graduate Scholarship, an FRQS doctoral training fellowship, and a McGill University Faculty of Medicine Studentship.
- G.B.L. is supported by scholarships from the FRQS, the CIHR, and Québec’s ministry of health and social services.
- Y.C. is supported by an FRQS doctoral training fellowship and the Lady Davis Institute/TD Bank Studentship Award.
- C-Y.S. is supported by a CIHR Canada Graduate Scholarship Doctoral Award, an FRQS doctoral training fellowship, and a Lady Davis Institute/ TD Bank Studentship Award.
Collapse
Affiliation(s)
- Satoshi Yoshiji
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada.
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada.
- Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Tianyuan Lu
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Guillaume Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Division of Infectious Diseases, McGill University Health Centre, Montréal, Québec, Canada
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julia Carrasco-Zanini-Sanchez
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Chen-Yang Su
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
| | - Yiheng Chen
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- 5 Prime Sciences, Montréal, Québec, Canada
| | - Kevin Liang
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
| | - Julian Daniel Sunday Willett
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
- Department of Anatomic Pathology and Laboratory Medicine, New York Presbyterian - Weill Cornell Medical Center, New York, NY, USA
| | | | - Darin Adra
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Yann Ilboudo
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Takayoshi Sasako
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Satoshi Koyama
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tetsushi Nakao
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Yossi Farjoun
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Fulcrum Genomics, Somerville, MA, USA
| | - Hugo Zeberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Sirui Zhou
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
| | - Michael Marks-Hultström
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rama Kaalia
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hesam Dashti
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, MA, USA
| | - Melina Claussnitzer
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jason Flannick
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Vincent Mooser
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
| | - Nicholas J Timpson
- Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - J Brent Richards
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada.
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada.
- Department of Twin Research, King's College London, London, UK.
| |
Collapse
|
20
|
Ballesteros-Pomar MD, Rodríguez-Urgellés E, Sastre-Belío M, Martín-Lorenzo A, Schnecke V, Segú L, Brosa M, Vilarrasa N. Assessment of the Potential Clinical and Economic Impact of Weight Loss in the Adult Population with Obesity and Associated Comorbidities in Spain. Adv Ther 2025; 42:1265-1282. [PMID: 39825974 PMCID: PMC11787177 DOI: 10.1007/s12325-024-03094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Obesity and its complications are associated with high morbidity/mortality and a significant healthcare cost burden in Spain. It is therefore essential to know the potential clinical and economic benefits of reducing obesity. The objective of this study is to predict the decrease in rates of onset of potential complications associated with obesity and the cost savings after a weight loss of 15% over 10 years in Spain. METHODS Data were combined in an adapted version of a weight loss benefit simulation model. Sources with demographic information on the Spanish population and the distribution of obesity and type 2 diabetes mellitus (T2DM) were used to obtain the data for the model. In addition, use was made of prevalence data on obesity-associated complications from a cohort of patients with obesity in the United Kingdom (UK). These data were combined by age and sex to create a Spanish synthetic cohort. RESULTS The simulation showed that, for a cohort of 100,000 individuals with a body mass index (BMI) of 30-50 kg/m2, a weight loss of 15% is estimated to lead to relevant relative risk reductions in obstructive sleep apnoea (OSA) (- 56.4%), T2DM (- 39.2%), asthma (- 20.2%) and arterial hypertension (- 18.7%). The estimated overall savings were €105 million for a cohort of 100,000 individuals, mainly resulting from the decrease in T2DM and arterial hypertension (23% and 22% of the total savings at year 10, respectively), as well as osteoarthritis and chronic kidney disease (CKD) (16% and 13%, respectively). CONCLUSIONS Sustained weight loss could significantly reduce the burden derived from future complications associated to obesity in Spain, as well as the excess economic cost associated with its treatment.
Collapse
Affiliation(s)
- María Dolores Ballesteros-Pomar
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León, Altos de Nava S/N, 24701, León, Spain
| | - Ened Rodríguez-Urgellés
- Market Access & Healthcare Consulting, Cencora PharmaLex Spain, C/del Comte d'Urgell, 240, 2D, 08036, Barcelona, Spain
| | - Miquel Sastre-Belío
- Market Access & Healthcare Consulting, Cencora PharmaLex Spain, C/del Comte d'Urgell, 240, 2D, 08036, Barcelona, Spain
| | - Alberto Martín-Lorenzo
- Department of Market Access and Public Affairs, Novo Nordisk, Vía de los Poblados 3, Edificio 6, Planta 3, 28033, Madrid, Spain.
| | - Volker Schnecke
- Real-World Science, Novo Nordisk, Østmarken 3A, 2860, Søborg, Denmark
| | - Lluís Segú
- Market Access & Healthcare Consulting, Cencora PharmaLex Spain, C/del Comte d'Urgell, 240, 2D, 08036, Barcelona, Spain
| | - Max Brosa
- Market Access & Healthcare Consulting, Cencora PharmaLex Spain, C/del Comte d'Urgell, 240, 2D, 08036, Barcelona, Spain
| | - Nuria Vilarrasa
- Department of Endocrinology and Nutrition, Hospital Universitari de Bellvitge-IDIBELL, C/de la Feixa Llarga S/N, 08907, Hospitalet de Llobregat, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta 0, 28029, Madrid, Spain
| |
Collapse
|
21
|
Młynarska E, Czarnik W, Dzieża N, Jędraszak W, Majchrowicz G, Prusinowski F, Stabrawa M, Rysz J, Franczyk B. Type 2 Diabetes Mellitus: New Pathogenetic Mechanisms, Treatment and the Most Important Complications. Int J Mol Sci 2025; 26:1094. [PMID: 39940862 PMCID: PMC11817707 DOI: 10.3390/ijms26031094] [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: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM), a prevalent chronic disease affecting over 400 million people globally, is driven by genetic and environmental factors. The pathogenesis involves insulin resistance and β-cell dysfunction, mediated by mechanisms such as the dedifferentiation of β-cells, mitochondrial dysfunction, and oxidative stress. Treatment should be based on non-pharmacological therapy. Strategies such as increased physical activity, dietary modifications, cognitive-behavioral therapy are important in maintaining normal glycemia. Advanced therapies, including SGLT2 inhibitors and GLP-1 receptor agonists, complement these treatments and offer solid glycemic control, weight control, and reduced cardiovascular risk. Complications of T2DM, such as diabetic kidney disease, retinopathy, and neuropathy, underscore the need for early diagnosis and comprehensive management to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Natasza Dzieża
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Jędraszak
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Gabriela Majchrowicz
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Filip Prusinowski
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Magdalena Stabrawa
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
22
|
Huang Z, Sun G, Li J, Zhang B, Lai G, Jing H, Zhou Y. Optimal exercise dose on Body Mass Index (BMI) in children and adolescents with overweight and obesity: a systematic review and bayesian model-based network meta-analysis. BMC Public Health 2025; 25:215. [PMID: 39827114 PMCID: PMC11742208 DOI: 10.1186/s12889-025-21405-3] [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: 09/05/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Exercise is widely used for obesity management, but the optimal doses of exercise for improving body mass index (BMI) in children and adolescents with overweight and obesity remain unclear. This study aimed to evaluate the dose‒response effects of various exercises on BMI in children and adolescents with overweight and obesity. METHODS A systematic search was conducted in Web of Science (Core Collection), PubMed/MEDLINE, Embase, Scopus, and the Cochrane Library for randomized controlled trials on relevant studies, covering literature up to July 2024. Three independent reviewers assessed bias via the Cochrane risk of bias tool. The quality of evidence was assessed using Confidence in Network Meta-Analysis framework. A dose‒response network meta-analysis was used to evaluate the impact of various exercise interventions and explore dose‒response relationships. All outcomes were analyzed with the mean difference (MD) and 95% credible intervals (CrIs) calculated for combined statistics. RESULTS The study included 39 publications with 1,814 participants, 47.3% female and a median age of 14 years. The intervention involved six exercise modalities. High-intensity interval training demonstrated a significant reduction in BMI (MD = -1.33, 95% CrIs - 2.01 to -0.66), followed by combined exercise (MD = -1.25, 95% CrIs - 1.93 to -0.61), moderate-intensity continuous training (MD = -1.09, 95% CrIs - 1.73 to -0.45), and mixed aerobic exercise (MD = -1.05, 95% CrIs - 1.67 to -0.42). There was an 'L'-shaped nonlinear dose-response relationship between total exercise dose and BMI, with 200 METs-min/day identified as the minimum exercise dose required to achieve a clinically meaningful reduction in BMI. CONCLUSION Low-quality evidence indicates that HIIT, CE, MAE, and MICT improve BMI in children and adolescents with overweight and obesity, with clarified exercise doses for clinical benefit. These findings are relevant for exercise prescription and public health policy. TRIAL REGISTRATION CRD42024566450.
Collapse
Affiliation(s)
- Zan Huang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Gang Sun
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Jiayu Li
- College of Education, Zhejiang Normal University, Jinhua, China
| | - Bin Zhang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
- Sports Coaching College, Beijing Sport University, Beijing, China
| | | | - Hongwei Jing
- School of Physical Education, Jiujiang University, Jiujiang, China
| | - Yulan Zhou
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
23
|
Daher Vianna AG, Alves DF, Assmann TS, Réa RR. The rise of weekly insulins: addressing the challenges of type 2 diabetes care in Brazil. Diabetol Metab Syndr 2025; 17:14. [PMID: 39810242 PMCID: PMC11734585 DOI: 10.1186/s13098-024-01560-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2D) is a global health concern with a rising prevalence, particularly in Brazil. Insulin therapy plays a crucial role in managing T2D, helping to maintain glucose and energy homeostasis. Moreover, early initiation of insulin is crucial for hyperglycemic control and prevention of chronic complications. Clinical guidelines recommend initiating insulin when other treatments fail. However, several barriers may delay its initiation, contributing to therapeutic inertia and patients' non-adherence. These barriers include fear of hypoglycemia, lack of adherence, the need for glucose monitoring, the injection method of insulin administration, social rejection associated with the stigma of injections, fear of weight gain, a sense of therapeutic failure at initiation, and lack of experience among some healthcare professionals. : In this context, the development of once-weekly insulin formulations could improve initial acceptance, adherence, treatment satisfaction, and consequently, the quality of life for patients. Currently, two once-weekly insulin treatments, insulin icodec and efsitora alfa, have shown promise in clinical trials, demonstrating efficacy and safety profiles similar or better than those of daily insulin therapies. SHORT CONCLUSION These once-weekly insulins have the potential to emerge as landmark achievements in the evolution of insulin therapy. This narrative review aims to evaluate the role of weekly insulins in managing T2D, providing insights into the potential benefits, challenges, and opportunities associated with a new weekly insulin therapy, specially within the Brazilian context.
Collapse
Affiliation(s)
- André Gustavo Daher Vianna
- Centro de Diabetes Curitiba, Department of Endocrine Diseases, Hospital Nossa Senhora das Graças, Curitiba, Brazil.
- Centro de Diabetes Curitiba, Rua Alcides Munhoz, 433 - 4º andar - Mercês, Curitiba, PR, 80810-040, Brazil.
| | | | | | - Rosângela Roginski Réa
- Serviço de Endocrinologia (SEMPR) do Hospital das Clínicas da Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| |
Collapse
|
24
|
Guo J, Mutailipu K, Wen X, Yin J, You H, Qu S, Chen H, Bu L. Association between lymphocyte to high-density lipoprotein cholesterol ratio and insulin resistance and metabolic syndrome in US adults: results from NHANES 2007-2018. Lipids Health Dis 2025; 24:9. [PMID: 39794792 PMCID: PMC11721163 DOI: 10.1186/s12944-024-02411-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) and metabolic syndrome (MetS) are significant global health challenges that increase the risk of various chronic diseases. The lymphocyte-to-high-density lipoprotein cholesterol ratio (LHR) has emerged as a novel inflammatory metabolic marker. The present study focused on evaluating the association between the LHR and both IR and MetS. METHODS We analyzed data from 14,779 adults aged ≥ 20 years from the National Health and Nutrition Examination Survey (2007-2018). To investigate the relationship between LHR and both IR and MetS, we conducted multivariable logistic regression analyses. The reliability of the results was validated through both stratified and sensitivity analyses. Furthermore, we thoroughly examined possible nonlinear associations by implementing a restricted cubic spline in conjunction with a threshold effect analysis. RESULTS Compared to the lowest LHR quartile, individuals in the highest quartile indicated significantly increased prevalence of IR (odds ratio = 3.72, 95% confidence intervals: 3.01-4.59) and MetS (odds ratio = 11.38, 95% confidence intervals: 8.85-14.63) in fully adjusted models. Subgroup analyses demonstrated that the association between the LHR and IR remained consistent across all subgroups, with no significant interaction effect observed. However, the association between LHR and MetS was more pronounced in female participants. Restricted cubic spline analyses revealed nonlinear associations between LHR and both IR and MetS. The threshold effect analyses identified inflection points at 0.055 for these non-linear relationships. CONCLUSIONS An elevated LHR was positively associated with the prevalence of IR and MetS, indicating its promising role in early screening and disease prevention through biological monitoring.
Collapse
Affiliation(s)
- Junwei Guo
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Kelibinuer Mutailipu
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xin Wen
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiajing Yin
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Hui You
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Shen Qu
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Haibing Chen
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Le Bu
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
25
|
Sakamoto K, Butera MA, Zhou C, Maurizi G, Chen B, Ling L, Shawkat A, Patlolla L, Thakker K, Calle V, Morgan DA, Rahmouni K, Schwartz GJ, Tahiri A, Buettner C. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab 2025; 37:121-137.e6. [PMID: 39437790 PMCID: PMC11711004 DOI: 10.1016/j.cmet.2024.09.012] [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: 01/09/2024] [Revised: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The mechanisms underlying obesity-induced insulin resistance remain incompletely understood, as impaired cellular insulin signaling, traditionally considered the primary driver of insulin resistance, does not always accompany impaired insulin action. Overnutrition rapidly increases plasma norepinephrine (NE), suggesting overactivation of the sympathetic nervous system (SNS). However, the role of the SNS in obesity is controversial, as both increased and decreased SNS activity (SNA) have been reported. Here, we show that reducing catecholamine (CA) release from the SNS protects against overnutrition-induced insulin resistance as well as hyperglucagonemia, adipose tissue dysfunction, and fatty liver disease, as we demonstrate utilizing a mouse model of inducible and peripherally restricted deletion of tyrosine hydroxylase (th; THΔper). A key mechanism through which heightened SNA induces insulin resistance is by triggering adipose tissue lipolysis. Increased SNA emerges as a critical driver in the pathogenesis of overnutrition-induced insulin resistance and metabolic disease independent of cellular insulin signaling.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary A Butera
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxue Zhou
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Maurizi
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bandy Chen
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Ling
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adham Shawkat
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Likhitha Patlolla
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kavira Thakker
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Victor Calle
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gary J Schwartz
- Department of Medicine & Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Azeddine Tahiri
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
Dhritlahre RK, Thakur N, Goel A, Patial V, Padwad Y, Saneja A. Self-Nanoemulsifying Formulation Improves Oral Bioavailability and Insulin Sensitizing Potency of Formononetin-Vitamin E Conjugate in Type 2 Diabetic Mice. Mol Pharm 2025; 22:255-269. [PMID: 39699518 DOI: 10.1021/acs.molpharmaceut.4c00886] [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] [Indexed: 12/20/2024]
Abstract
The escalating incidence of obesity, diabetes, and insulin resistance has become a significant global health concern. In this study, we have developed a self-nanoemulsifying delivery system (SNEDS) of formononetin-vitamin E conjugate (VESylated-FMN) for improving its oral bioavailability and improving insulin sensitivity and glycemic control. The developed SNEDS were characterized using dynamic light scattering and transmission electron microscopy. Thereafter, the loading capacity, in vitro release, thermodynamic, and gastrointestinal stability of the developed formulation were evaluated. The safety and oral bioavailability of VESylated-FMN-SNEDS were assessed in Sprague-Dawley rats, whereas insulin-sensitizing potency was assessed in high-fat diet-induced type 2 diabetic mice. The VESylated-FMN-SNEDS quickly emulsified on dilution (droplet size ∼79.17 nm) and showed remarkable thermodynamic and gastrointestinal stability. The developed formulation demonstrated enhanced oral bioavailability (∼1.3-fold higher AUC0-t) of VESylated-FMN without liver and kidney injury. Consequently, VESylated-FMN-SNEDS significantly improves insulin sensitivity and glycemic control in HFD-fed mice compared to VESylated-FMN by upregulating the transcript level of insulin-sensitizing genes. Therefore, the SNEDS formulation could be an effective strategy to augment the oral bioavailability and insulin-sensitizing potency of VESylated-FMN.
Collapse
Affiliation(s)
- Rakesh Kumar Dhritlahre
- Formulation Laboratory, Dietetics & Nutrition Technology Division, CSIR - Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Navneet Thakur
- Formulation Laboratory, Dietetics & Nutrition Technology Division, CSIR - Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
| | - Abhishek Goel
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR - Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
| | - Vikram Patial
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR - Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
| | - Yogendra Padwad
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR - Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
| | - Ankit Saneja
- Formulation Laboratory, Dietetics & Nutrition Technology Division, CSIR - Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
27
|
Ribeiro T, Reis M, Vasconcelos V, Urbatzka R. Phenotypic screening in zebrafish larvae identifies promising cyanobacterial strains and pheophorbide a as insulin mimetics. Sci Rep 2024; 14:32142. [PMID: 39739113 PMCID: PMC11685485 DOI: 10.1038/s41598-024-83986-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
Diabetes is a pandemic disease that causes the loss of control of glucose regulation in the organism, in consequence of dysfunction of insulin production or functionality. In this work, the antidiabetic bioactivity of 182 fractions from 19 cyanobacteria strains derived from the LEGE Culture Collection were analysed using the 2-NBDG assay in zebrafish larvae. From this initial screening, two fractions (57 (06104_D) and 107 (03283_B)) were identified as promising insulin mimetics. These were further characterized by measuring glucose levels in whole larvae, the expression of glucose transporters (GLUT 1-3) using western blot, and the mRNA expression levels of the glut2, pepck, and insa genes using real-time qPCR. Both fractions showed a decrease in free glucose levels. Furthermore, exposure to fraction 06104_D decreased GLUT1 and increased insa mRNA levels. The chemical composition of these fractions was determined using LC-HRESIMS/MS and compared to inactive fractions of the same polarity in order to identify the unique bioactive molecules. The molecular networks constructed using the GNPS platform revealed that fraction 06104_D contained mass clusters primarily composed of chlorins, lipids, and terpenoids, while fraction 03283_B contained xanthophylls, peptides, and terpenoids. To correlate the observed activity with the chemical composition of fraction 06104_D, pheophorbide a was chosen as a representative of chlorophyll derivatives. Exposure to zebrafish larvae at 10 and 20 µM confirmed the increased glucose uptake on the 2-NBDG assay. These findings highlight the bioactivity of chlorophyll derivatives as insulin mimetic compounds, as well as cyanobacteria as a source of potential therapeutic diabetes applications.
Collapse
Affiliation(s)
- Tiago Ribeiro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, Matosinhos, 4450-208, Portugal.
- Faculty of Sciences, University of Porto, Rua do Campo Alegre, Porto, 1021, 4169-007, Portugal.
| | - Mariana Reis
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, Matosinhos, 4450-208, Portugal
| | - Vitor Vasconcelos
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, Matosinhos, 4450-208, Portugal
- Faculty of Sciences, University of Porto, Rua do Campo Alegre, Porto, 1021, 4169-007, Portugal
| | - Ralph Urbatzka
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, Matosinhos, 4450-208, Portugal
| |
Collapse
|
28
|
Glendinning JI, Williams N. Fighting Fire with Fire: Impact of Sugary Diets on Metabolically Deranged Mice. Nutrients 2024; 17:100. [PMID: 39796534 PMCID: PMC11722652 DOI: 10.3390/nu17010100] [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: 11/15/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: There is controversy about the health risks of sugary diets. A recent study reported that chronic consumption of 11% sugar solutions improved glycemic control in lean mice. Based on this finding, we hypothesized that chronic consumption of the same 11% sugar solutions would also improve glycemic control in metabolically deranged mice. Methods: We exposed mice to a high-fat/high-sugar diet for 12 weeks. Then, we switched the mice to a control (i.e., standard chow) or one of four experimental diets for 8 weeks. The experimental diets contained standard chow plus an 11% solution of glucose or high-fructose syrup. The sugar syrups were derived from corn or cellulose. We included the cellulosic syrups because they contain polyphenols, which are thought to promote glycemic control. We measured body weight, adiposity, glucose tolerance, insulinemia, insulin sensitivity, body composition, and avidity for sweeteners. Results: Mice switched to the control diet lost weight, whereas mice switched to the experimental diets remained obese and hyperinsulinemic. Thus, the experimental diets did not cause the mice to regain normal metabolic health. Nevertheless, we observed (i) improvements in glucose tolerance in mice on both the control and experimental diets; (ii) reduced insulinemia and enhanced insulin sensitivity in mice offered the cellulosic syrups; (iii) elevations in cephalic-phase insulin responses in mice on the experimental diets; and (iv) increased avidity for sweeteners in mice on the control but not the experimental diets. Conclusions: Switching metabolically deranged mice to the experimental diets, particularly those with cellulosic sugars, improved glucose tolerance and insulin sensitivity.
Collapse
Affiliation(s)
- John I. Glendinning
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA;
| | - Niki Williams
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA;
| |
Collapse
|
29
|
Sun Q, Wang J, Wang H, Yu H, Wan K, Ma F, Wang R. Effect of Long-Term Taurine Supplementation on the Lipid and Glycaemic Profile in Adults with Overweight or Obesity: A Systematic Review and Meta-Analysis. Nutrients 2024; 17:55. [PMID: 39796489 PMCID: PMC11722866 DOI: 10.3390/nu17010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Taurine has been demonstrated to regulate and improve metabolic health. However, physiological and pathological differences among individuals with overweight or obesity may result in varied responses to taurine supplementation. This study aims to estimate the effects of long-term taurine supplementation on blood lipids, glycemia, and insulin sensitivity in adults with overweight or obesity through a systematic review and meta-analysis. METHODS The literature search was based on six databases (Web of Science, PubMed, Scopus, EMBASE, Cochrane, and SPORTDiscus) up to October 2024. Subgroup analyses were performed based on daily taurine intake dosage (<3 g or 3 g), overweight (BMI 25-29.9 kg/m2), and obesity (BMI ≥30 kg/m2). RESULTS The final number of studies that met the inclusion criteria was 9 RCTs. The overall analysis showed that taurine supplementation significantly decreased TG (WMD = -0.56 mg/dL, 95% CI: -0.92 to -0.2, p = 0.002, I2 = 63%), TC (WMD = -0.71 mg/dL, 95% CI: -1.17 to -0.25, p = 0.002, I2 = 73%), and fasting insulin (WMD = -2.15 µU/mL, 95% CI: -3.24 to -1.06, p = 0.0001, I2 = 9%). In the subgroup analysis, long-term taurine intake led to BMI improvement in overweight adults (WMD = -1.14 kg/m2, 95% CI: -1.81 to -0.47, p = 0.0008, I2 = 0%). Meanwhile, improvements in HbA1c (WMD = -0.33%, 95% CI: -0.53 to -0.12, p = 0.002, I2 = 16%) and HOMA-IR (WMD = -0.91, 95% CI: -1.74 to -0.08, p = 0.003, I2 = 54%) were observed only in obese participants following taurine supplementation. Additionally, the long-term intake of 3 g of taurine significantly improved HbA1c (WMD = -0.37%, 95% CI: -0.61 to -0.13, p = 0.003, I2 = 0%) and FPG levels (WMD = -7.14 mg/dL, 95% CI: -12.53 to -1.74, p = 0.003, I2 = 70%) in overweight/obesity. CONCLUSIONS Long-term taurine supplementation is particularly effective in improving glycemic control and insulin sensitivity in obesity. Furthermore, higher doses of taurine (3 g per day) demonstrate even greater improvements in glycemic control.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Q.S.); (J.W.); (H.W.); (H.Y.); (K.W.); (F.M.)
| |
Collapse
|
30
|
Shukla A, Meena K, Gupta A, Sandhir R. 1H NMR-Based Metabolomic Signatures in Rodent Models of Sporadic Alzheimer's Disease and Metabolic Disorders. ACS Chem Neurosci 2024; 15:4478-4499. [PMID: 39629865 DOI: 10.1021/acschemneuro.4c00510] [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] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological disorder that impacts the elderly population all over the globe. Evidence suggests association between AD and metabolic disorders such as diabetes mellitus (DM) and obesity (OB). The present study is an attempt to evaluate metabolic alterations in the serum and brain through NMR spectroscopy with the aim to identify shared metabolic signatures. AD was induced in rats by stereotactic intracerebroventricular injection of oligomerized Aβ-42 peptide into the brain. DM and OB were induced by intraperitoneal injection of streptozotocin and feeding rats on a high-fat diet, respectively. The metabolic alterations obtained through 1H NMR spectroscopy were further subjected to multivariate analysis by principal component analysis and partial least-squares discrimination for identification of metabolic signatures. In the serum, the levels of lactate and betaine were increased in AD, DM, and OB rats. On the other hand, the metabolite profile of brain indicated increase in the levels of lactate, N-acetylaspartate, and creatinine in AD, DM, and OB rats. Additionally, the concentration of neurochemicals such as glutamate, GABA, N-acetylglutamate, and myo-inositol were also elevated. The alterations in neurotransmitters and cerebral energy metabolism were accompanied by deficits in cognition assessed by Morris water maze in AD, DM, and OB rats. The perturbed metabolic profiles were accompanied by the presence of pathogenic amyloid deposits visualized by Congo red stain in the brains of AD, DM, and OB rats. Overall, the study identifies common metabolic signatures in AD, DM, and OB that may be involved in etiopathogenesis and also suggests linkages between these three conditions.
Collapse
Affiliation(s)
- Ananya Shukla
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| | - Khushbhu Meena
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Ashish Gupta
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Rajat Sandhir
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| |
Collapse
|
31
|
Chen X, Lin E, Haghighatian MM, Shepard LW, Hattar S, Kuruvilla R, Zhao H. Light modulates glucose and lipid homeostasis via the sympathetic nervous system. SCIENCE ADVANCES 2024; 10:eadp3284. [PMID: 39661675 PMCID: PMC11633741 DOI: 10.1126/sciadv.adp3284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Light is an important environmental factor for vision and for diverse physiological and psychological functions. Light can also modulate glucose metabolism. Here, we show that in mice, light is critical for glucose and lipid homeostasis by regulating the sympathetic nervous system, independent of circadian disruption. Light deprivation from birth elicits insulin hypersecretion, glucagon hyposecretion, lower gluconeogenesis, and reduced lipolysis by 6 to 8 weeks in male, but not female, mice. These metabolic defects are consistent with blunted sympathetic activity, and indeed, sympathetic responses to a cold stimulus are substantially attenuated in dark-reared mice. Further, long-term dark rearing leads to body weight gain, insulin resistance, and glucose intolerance. Notably, metabolic dysfunction can be partially alleviated by 5 weeks exposure to a regular light-dark cycle. These studies provide insight into circadian-independent mechanisms by which light directly influences whole-body physiology and better understanding of metabolic disorders linked to aberrant environmental light conditions.
Collapse
Affiliation(s)
- Xiangning Chen
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eugene Lin
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
32
|
Amer J, Amleh A, Salhab A, Kolodny Y, Yochelis S, Saffouri B, Paltiel Y, Safadi R. High-fat diet mouse model receiving L-glucose supplementations propagates liver injury. Front Nutr 2024; 11:1469952. [PMID: 39742098 PMCID: PMC11687001 DOI: 10.3389/fnut.2024.1469952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/11/2024] [Indexed: 01/03/2025] Open
Abstract
Background and aims Limited data link manufactured sweeteners impact on metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to evaluate the effects of manufactured sugars (L-glucose) compared to natural sugars (D-glucose) on phenotype, molecular and metabolic changes in mice models fed with either regular diet (RD) or high fat diet (HFD). Methods C57BL/6 mice fed 16-weeks with either RD; 70% carbohydrate or HFD; 60% fat, with or without additional glucose (Glu, at 18% w/v) to drinking tap water at weeks 8-16; of either natural (D-Glu) or manufactured (L-Glu) sugars. Liver inflammation (ALT and AST serum levels, liver H&E histologic stains and cell viability profile by p-AKT), liver fibrosis [quantitated α smooth-muscle-actin (αSMA) by western blot and RT-PCR, Masson Trichrome staining (MTC) of liver tissue], liver lipid [steatosis stain by H&E, Adipose Differentiation-Related Protein (ADRP) lipid accumulation, serum and lipid peroxidation Malondialdehyde (MDA) markers by ELISA], glucose hemostasis (serum Glucose and C-peptide with HOMA-IR score calculation) and liver aspects [hepatic glucose transporter 2 (GLUT2), insulin receptor (IR) expressions and GYS2/PYGL ratio] evaluated. Results D- and L-Glu supplementations propagate hepatocytes ballooning and steatosis in HFD-fed mice and were associated with αSMA down-expressions by 1.5-fold compared to the untreated group while showed an acceleration in liver fibrosis in the RD-fed mice. Lipid profile (Steatosis, ADRP and MDA) significantly increased in HFD-fed mice, both Glu supplementations (mainly the L-Glu) increased serum MDA while decreased ADRP. HOMA-IR score and IR significantly increased in HFD-fed mice, with further elevation in HOMA-IR score following Glu supplementations (mainly L-Glu). The increase in HOMA-IR negatively correlated with IR and Glut2 expressions. D- and L-Glu supplementations showed significant decrease of Glycogenesis (low GYS2/PYGL ratio) and unchanged p-AKT pattern compared to their RD counterparts. Conclusion Our data indicate an increase in rate of de-novo lipogenesis (DNL) in RD-fed mice (High carbohydrate diet) and liver fibrosis following additional sugar supplementations. In contrast, HFD-fed mice (with pre-existing high lipid profile) supplemented with sugar showed less liver fibrosis, because of reduced de-novo fatty acids synthesis and subsequently, the lipid oxidation pathways become dominated and induce the net results of lipid clearance.
Collapse
Affiliation(s)
- Johnny Amer
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Athar Amleh
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Ahmad Salhab
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Yuval Kolodny
- Applied Physics Department, Center for Nanoscience and Nanotechnology, Hebrew University Givaat Ram, Jerusalem, Israel
| | - Shira Yochelis
- Applied Physics Department, Center for Nanoscience and Nanotechnology, Hebrew University Givaat Ram, Jerusalem, Israel
| | - Baker Saffouri
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Yossi Paltiel
- Applied Physics Department, Center for Nanoscience and Nanotechnology, Hebrew University Givaat Ram, Jerusalem, Israel
| | - Rifaat Safadi
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| |
Collapse
|
33
|
Zhang L, Lou K, Zhang Y, Leng Y, Huang Y, Liao X, Liu X, Feng S, Feng G. Tools for regulating metabolic diseases: extracellular vesicles from adipose macrophages. Front Endocrinol (Lausanne) 2024; 15:1510712. [PMID: 39735643 PMCID: PMC11674605 DOI: 10.3389/fendo.2024.1510712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/31/2024] Open
Abstract
Metabolic diseases have gradually become one of the most significant global medical burdens. Diseases such as obesity, diabetes, and metabolic syndrome, along with their complications, are clinically categorized as metabolic diseases. Long-term oral medication significantly reduces patient compliance and quality of life. Therefore, alternative therapies that intervene at the cellular level or target the root causes of metabolic diseases might help change this predicament. Research has found that extracellular vesicles derived from adipose macrophages can effectively regulate metabolic diseases by influencing the disease's development. This regulation is likely related to the role of these extracellular vesicles as important mediators in modulating adipose tissue function and insulin sensitivity, and their involvement in the crosstalk between adipocytes and macrophages. This review aims to describe the regulation of metabolic diseases mediated by adipose macrophage-derived extracellular vesicles, with a focus on their involvement in adipocyte crosstalk, the regulation of metabolism-related autoimmunity, and their potential as therapeutic agents for metabolic diseases, providing new avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| | - Kecheng Lou
- Department of Urology, Lanxi People’s Hospital, Jinhua, Zhejiang, China
| | - Yunmeng Zhang
- Department of Anesthesiology, Jiujiang College Hospital, Jiujiang, Jiangxi, China
| | - Yuanjing Leng
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| | - Yuqing Huang
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| | - Xinxin Liao
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| | - Xiaoliang Liu
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| | - Shangzhi Feng
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| | - Guoqiang Feng
- Department of Rehabilitation, Jiujiang College Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
34
|
Aly O, Mekky RH, Pereira F, Diab YM, Tammam MA, El-Demerdash A. Deciphering the potential of Cymbopogon citratus (DC.) Stapf as an anti-obesity agent: phytochemical profiling, in vivo evaluations and molecular docking studies. Food Funct 2024; 15:12146-12168. [PMID: 39585680 DOI: 10.1039/d4fo04602a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Based on its anti-inflammatory and antioxidant properties, Cymbopogon citratus (DC) Stapf is commonly used in traditional and modern medicine to cure different diseases. The present study investigates the potential of C. citratus organic extract as an anti-obesity drug in a HCHFD (high-carbohydrate, high-fat diet) model for obese rats. Its negative hypolipidemic effect has been confirmed through biochemical and histological methods. Fifty male albino rats were randomly divided into five groups (10 rats each) Group I (Control group), Group II (HCHFD group), Group III (C. citratus group), Group IV (HCHFD + C. citratus group) and Group V (HCHFD + Orlistat group). Serum glucose levels and lipid profiles were quantified using a spectrophotometer. Insulin, apelin, and adiponectin parameters were measured using ELISA (enzyme-linked immunosorbent assay) kits, while real-time PCR following extraction and purification was used for apelin, apelin receptor genes (APJ), and adiponectin gene expression evaluation. Besides, C. citratus methanolic extract was subjected to untargeted metabolic profiling via RP-HPLC-QTOF-MS and MS/MS, disclosing the presence of 52 secondary metabolites where they mainly belonged to phenolic compounds viz., flavones and hydroxycinnamic acids, among other metabolites with predominance of derivatives of luteolin and O-coumaroyl-O-feruloylglycerol. Our findings were further strengthened by computational-based virtual screening protocols that included molecular docking (MDock) and Structure-Activity Relationships (SARs). The MDock studies revealed that the three main flavone-containing metabolites, each with a luteolin C6-glycosylation core featuring two sugar units (16, 25, and 31), outperformed the positive control (8EH, a triazole derivative) known to bind to the APJ protein. These metabolites exhibited exceptional binding affinities, with estimated free binding energy (ΔGB) values of -9 kcal mol-1 or lower, likely due to potential hydrogen bond interactions with the Arg168 residue of the APJ protein. Additionally, the pharmacokinetic, physicochemical, and toxicity profiles of the 11 major metabolites from C. citratus leaf extract were assessed, revealing a profile like that of the positive control in the three selected flavone metabolites. Based on the acquired data, it can be concluded that C. citratus shows strong potential as a hypolipidemic agent and could play a significant role in managing obesity and mitigating its associated complications.
Collapse
Affiliation(s)
- Omnia Aly
- Department of Medical Biochemistry, National Research Centre, Cairo 12622, Egypt.
| | - Reham Hassan Mekky
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, 11829, Cairo, Egypt
| | - Florbela Pereira
- LAQV REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829516 Caparica, Portugal
| | - Yasser M Diab
- Department of Biochemistry, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt
| | - Mohamed A Tammam
- Department of Biochemistry, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt
| | - Amr El-Demerdash
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
- Faculty of Sciences, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
35
|
Lee WJ, Cho KJ, Kim GW. Mitigation of Atherosclerotic Vascular Damage and Cognitive Improvement Through Mesenchymal Stem Cells in an Alzheimer's Disease Mouse Model. Int J Mol Sci 2024; 25:13210. [PMID: 39684920 DOI: 10.3390/ijms252313210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by progressive memory loss and other cognitive disturbances. Patients with AD can be vulnerable to vascular damage, and damaged vessels can lead to cognitive impairment. Mesenchymal stem cell (MSC) treatment has shown potential in ameliorating AD pathogenesis, but its effect on vascular function remains unclear. This study aimed to improve cognitive function by alleviating atherosclerosis-induced vessel damage using MSCs in mice with a genetic AD background. In this study, a 5xFAD mouse model of AD was used, and atherosclerotic vessel damage was induced by high-fat diets (HFDs). MSCs were injected into the tail vein along with mannitol in 5xFAD mice on an HFD. MSCs were detected in the brain, and vascular damage was improved following MSC treatment. Behavioral tests showed that MSCs enhanced cognitive function, as measured by the Y-maze and passive avoidance tests. Additionally, muscle strength measured by the rotarod test was also increased by MSCs in AD mice with vessel damage induced by HFDs. Overall, our results suggest that stem cells can alleviate vascular damage caused by metabolic diseases, including HFDs, and vascular disease in individuals carrying the AD gene. Consequently, this alleviates cognitive decline related to vascular dementia symptoms.
Collapse
Affiliation(s)
- Woong Jin Lee
- Department of Neurology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyoung Joo Cho
- Department of Life Science, Kyonggi University, Suwon 16227, Republic of Korea
| | - Gyung Whan Kim
- Department of Neurology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
36
|
Tyczyńska M, Hunek G, Kawecka W, Brachet A, Gędek M, Kulczycka K, Czarnek K, Flieger J, Baj J. Association Between Serum Concentrations of (Certain) Metals and Type 2 Diabetes Mellitus. J Clin Med 2024; 13:7443. [PMID: 39685901 DOI: 10.3390/jcm13237443] [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: 10/28/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
The findings regarding trace element concentrations in patients diagnosed with type 2 diabetes and healthy controls are inconsistent, and therefore, we determined to gather them in the form of a review to further indicate the need for more advanced knowledge development. In our study, we reviewed articles and studies that involved the topics of micronutrient and metal associations with the occurrence and development of type 2 diabetes. We mainly included works regarding human-based studies, but with limited research results, animal-based research was also taken into account. With some newer studies, we reached for initial assumptions of previous statements. The results indicated that higher serum levels of lead, cadmium, arsenic, bromine, barium, strontium, nickel, aluminum, calcium, copper, and ferritin are positively associated with diabetic prevalence. Both too-low and too-high levels of zinc, selenium, and magnesium may be connected to the development of diabetes. Chromium has the capability of insulin response modulation, with enhanced insulin-cell binding, and thus, lower serum levels of chromium can be found in diabetic patients. There are contradictory discoveries regarding manganese. Its supplementation can possibly cease the development of insulin resistance and type 2 diabetes. On the contrary, other studies reported that there is no such connection. Our work indicates that, as micronutrients play a significant role in the pathogenesis of metabolic disorders, more research regarding their bodily homeostasis and type 2 diabetes should be conducted.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Gabriela Hunek
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Weronika Kawecka
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Adam Brachet
- Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Marta Gędek
- Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Kinga Kulczycka
- Institute of Medical Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1, 20-708 Lublin, Poland
| | - Katarzyna Czarnek
- Institute of Medical Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1, 20-708 Lublin, Poland
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| |
Collapse
|
37
|
Ma L, Wang Z, Sun L, Li M, Wu Q, Liu M, Xu M, Shi G, Yin J, Wang Y, Xu L. Association analysis between serum asprosin and metabolic characteristics, Complications in type 2 diabetic patients with different durations. J Diabetes Investig 2024; 15:1781-1787. [PMID: 39275947 PMCID: PMC11615684 DOI: 10.1111/jdi.14313] [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: 05/15/2024] [Revised: 08/08/2024] [Accepted: 08/31/2024] [Indexed: 09/16/2024] Open
Abstract
AIMS/INTRODUCTION To investigated the association between serum asprosin and metabolic characteristics in type 2 diabetes mellitus patients with different durations. MATERIALS AND METHODS A total of 436 patients with type 2 diabetes mellitus were enrolled in this study from the community health service center in southeastern Shanxi Province. All the patients were divided into two groups according to their diabetes duration: diabetes duration ≤5 years group (n = 132) and diabetes duration ≥10 years group (n = 304). Fasting blood samples were gathered and serum asprosin was tested. Pearson/Spearman correlation analysis was carried out. RESULTS Asprosin was comparable between the two groups. Asprosin was positively correlated with systolic blood pressure (SBP), triglycerides, creatinine, serum uric acid and low-density lipoprotein cholesterol in the diabetes duration ≤5 years group (P < 0.05). In the diabetes duration ≥10 years group, asprosin was independently correlated with SBP, diastolic blood pressure, body mass index, total cholesterol, triglycerides, low-density lipoprotein cholesterol, creatinine, serum uric acid, fasting plasma glucose and glycosylated hemoglobin (P < 0.05). Asprosin was associated with alanine aminotransferase and estimated glomerular filtration rate (P < 0.05). Multiple linear regression analysis found that SBP and diastolic blood pressure is an independent factor related to serum asprosin in the group with diabetes duration ≤5 years (P < 0.05). Fasting plasma glucose, SBP, total cholesterol and serum uric acid is an independent factor related to serum asprosin in the group with diabetes duration ≥10 years (P < 0.05). CONCLUSIONS Serum asprosin was significantly increased in the group with diabetes duration ≥10 years, and glycosylated hemoglobin, blood pressure and estimated glomerular filtration rate were independent risk factors in long-duration type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Lijing Ma
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| | - Zhengqian Wang
- First Clinical Medical CollegeShanxi Medical UniversityTaiyuanShanxiChina
| | - Li Sun
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| | - Mina Li
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| | - Qianqian Wu
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| | - Ming Liu
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| | - Minggang Xu
- Department of EndocrinologyChangzhi Second People's HospitalChangzhiChina
| | - Guoliang Shi
- Department of EndocrinologyChangzhi Second People's HospitalChangzhiChina
| | - Jianhong Yin
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| | - Yan Wang
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
- Clinical Research Center For Endocrine and Metabolic Diseases of Shanxi Medical UniversityTaiyuanShanxiChina
- Shanxi Innovation Center for Integrated Management of Hypertension, Hyperlipidemia and Hyperglycemia Correlated with Cardiovascular and Cerebrovascular DiseasesTaiyuanShanxiChina
| | - Linxin Xu
- Department of Endocrinology, First Hospital of Shanxi Medical UniversityShanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
38
|
Song H, Lu J, Chu Q. Polyphenols from Prunus salicina L. alleviate weight gain, obesity-related hyperlipidemia, hepatic steatosis, hyperglycemia, and modulate gut microbiota in mice fed a high-fat diet. Nutr Res 2024; 132:152-163. [PMID: 39580918 DOI: 10.1016/j.nutres.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
Hyperlipidemia, hepatic steatosis, and hyperglycemia are prevalent metabolic disorders closely linked to obesity. The objective of this research was to examine the potential advantageous impacts of polyphenols extracted from Prunus salicina L. fruit (PSFP) on hyperlipidemia, hepatic steatosis, and hyperglycemia induced by a high-fat diet (HFD), as well as to elucidate the underlying mechanisms involved. Male C57BL/6J mice, free from specific pathogens, were assigned randomly into three groups. These groups were then subjected to a 14-week dietary intervention, including a low-fat diet, an HFD, or an HFD plus with PSFP via intragastric administration. The obesity-related biochemical indexes were evaluated. To assess alterations in gut microbiota resulting from PSFP treatment, 16S rRNA sequencing was performed. UPLC-ESI-MS/MS assay identified 162 distinct polyphenolic compounds in PSFP. The administration of PSFP significantly reduced both body weight gain and hyperlipidemia induced by HFD. In addition, PSFP ameliorated hepatic steatosis induced by HFD and enhanced liver function in mice. PSFP treatment also ameliorated HFD-induced insulin resistance and hyperglycemia, evidenced by the observed decrease in fasting serum concentrations of glucose and insulin, improved insulin sensitivity, and restored glucose tolerance. Moreover, PSFP modulated the composition and abundance of specific microbial genus, including Lachnospiraceae NK4A136 group, Akkermansia, Parabacteroides, Enterococcus, Adlercreutzia, and Roseburia. Correlation analysis indicated significant associations between gut microbiota and physiological indices associated with obesity. These findings suggested that PSFP supplementation ameliorated HFD-induced hyperlipidemia, hepatic steatosis, and hyperglycemia, potentially through modulating the gut microbiota composition and abundance of specific taxa.
Collapse
Affiliation(s)
- Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, PR China.
| | - Jing Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, PR China
| | - Qiang Chu
- College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
39
|
Zhang L, Luo P, Li H, Pan Y, Zhang H, Si X, Chen W, Huang Y. Chicken GLUT4 function via enhancing mitochondrial oxidative phosphorylation and inhibiting ribosome pathway in skeletal muscle satellite cells. Poult Sci 2024; 103:104403. [PMID: 39515116 PMCID: PMC11584589 DOI: 10.1016/j.psj.2024.104403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
Glucose Transporter 4 (GLUT4) is a crucial protein facilitating glucose uptake and metabolism across cell membranes in mammals. However, information on GLUT4 in birds has historically been limited. In this study, we investigated the dynamic expression profile of chicken GLUT4 using real-time quantitative PCR (RT-qPCR) and examined its potential effects and mechanisms via GLUT4 overexpression and RNA sequencing (RNA-seq) in chicken primary skeletal muscle satellite cells (CP-SMSCs). Our results demonstrated that chicken GLUT4 is differentially expressed across tissues, with predominant expression in skeletal muscles, and across developmental stages of CP-SMSCs, with notable upregulation during the phases of cell proliferation and early differentiation. Notably, 0.1 μM insulin for 60 min significantly elevated the expression of GLUT4 in CP-SMSCs (P < 0.05). GLUT4 overexpression in CP-SMSCs promoted cell proliferation, as evidenced by Cell Counting Kit-8 (CCK-8) (P < 0.05) and 5-Ethynyl-2'-Deoxyuridine (EDU) assays (P < 0.05), and enhanced glucose consumption following 0.1 μM insulin treatment (P < 0.05). However, it inhibited glucose consumption 12 h after the addition of 5 g/L glucose (P < 0.05). After overexpressing GLUT4, we identified 302 differentially expressed genes (DEGs) in CP-SMSCs, with 134 upregulated and 168 downregulated. These DEGs are primarily enriched in pathways such as oxidative phosphorylation, ribosome, cardiac muscle contraction, ATP metabolic processes, and mitochondrial protein complexes. Specifically, in the enriched oxidative phosphorylation pathway, the upregulated DEGs (12) encode mitochondrial proteins, while the downregulated DEGs (6) are nuclear genome-derived. The ribosomal pathway is predominantly inhibited, accompanying with the downregulation of the translocase of outer mitochondrial membrane 7 (TOMM7)/translocase of inner mitochondrial membrane 8 (TIMM8A) complex responsible for mitochondrial protein transport, and a reduction in 28S (LOC121106978) and 18S (LOC112533601) ribosomal rRNAs. In conclusion, chicken GLUT4 is dynamically modulated during development and acts as an insulin responder that significantly regulates cellular glucose uptake and cell proliferation. This regulation occurs mainly through enhancing the mitochondrial oxidative phosphorylation and inhibiting ribosomal pathway.
Collapse
Affiliation(s)
- Lin Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Pengna Luo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Huihong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Yuxian Pan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Huaiyong Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Xuemeng Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Wen Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Yanqun Huang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China.
| |
Collapse
|
40
|
Huang W, Huang GP, Zhang LX, da Yu E, Yang WK, Ye M, Zou SQ, Ni L, He HQ. Lignan-rich extract from Cinnamomum camphora leaf attenuates metabolic syndrome by modulating glycolipid metabolism and gut microbiota in T2DM mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156118. [PMID: 39489989 DOI: 10.1016/j.phymed.2024.156118] [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: 02/28/2024] [Revised: 05/31/2024] [Accepted: 07/04/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a serious metabolic syndrome with high mortality and disability rates globally, which usually caused by unhealthy dietary patterns. Cinnamomum camphora leaf is a traditional Chinese medicinal herb used for attenuating hyperglycemia and digestive disorder, and high level of lignans has been found in C. camphora leaf. PURPOSE This study aimed to examine the chemical composition of lignans extracted from C. camphora leaf (LCCL), and illustrate its therapeutic effect and mechanism on T2DM and its concomitant glycolipid metabolic disorder. METHODS The components of LCCL were separated and purified by silica gel and macroporous adsorption resin, and were distinguished through LC/MS and NMR. The antioxidant activity of LCCL was determined by free radical scavenging assay in vitro; the hypoglycemic and hypolipidemic abilities were evaluated by α-glucosidase, α-amylase and pancreatic lipase inhibition trials, respectively. T2DM model mice were established by high-sugar and high-fat (HSHF) feed together with streptozotocin (STZ) infection, and then grouped to assess the effect of LCCL treatment. Hematoxylin-eosin (H&E), Periodic Acid-Schiff (PAS) and oil red O staining were employed to analyze the histopathology. qRT-PCR assay, 16S rRNA analysis, and western blot were conducted to illuminate the anti-diabetic mechanism of LCCL. RESULTS 6 sesamin lignans were identifed from LCCL. The in vitro assays showed strong inhibitive abilities of LCCL with low IC50 on DPPH (33.68 ± 0.54 μg/ml),O2- (39.25 ± 0.61 μg/ml), OH• (45.72 ± 0.72 μg/ml), α-glucosidase (0.82 ± 0.14 mg/ml), α-amylase (0.86 ± 0.11 mg/ml) and pancreatic lipase (0.91 ± 0.12 mg/ml). LCCL treatment (100, 200 and 400 g kg-1mg kg-1) gradually decreased the fasting blood glucose (FBG) and fasting insulin (FINS), improved the glucose and insulin tolerance, down-regulated the homeostasis model assessment insulin resistance (HOMA-IR) indexes, alleviated the hepatic inflammatory response and oxidative stress, promoted the glycogen storage and depleted the fat accumulation in the liver. Besides, LCCL administration alleviated the glycolipid metabolism disorder in T2DM mice with a gut microbiota dependent manner, that significantly increased biodiversity, altered the composition of gut microbiota and increased the proportion of Lactobacillus. CONCLUSION The lignan-rich extract of C. camphor leaf (LCCL), containing at least 6 lignans compounds, displayed promising antioxidant, hypoglycemic and hypolipidemic activities. The treatment of LCCL alleviated the glycolipid metabolism disorder in T2DM mice with a gut microbiota dependent manner. These finding suggested that LCCL should be further investigated to develop its complementary therapeutic effect on T2DM.
Collapse
Affiliation(s)
- Wei Huang
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, China.
| | - Guan-Peng Huang
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of JunCao Science and Ecology (College of Carbon Neutrality), Fujian Agriculture and Forestry University, China
| | - Lan-Xiong Zhang
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, China; College of Plant Protection, Fujian Agriculture and Forestry University, China
| | - En da Yu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wang-Ke Yang
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Min Ye
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China
| | - Shuang-Quan Zou
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, China
| | - Lin Ni
- Engineering Research Institute of Conservation, Utilization of Natural Bioresources, Fujian Agriculture and Forestry University, China; College of Plant Protection, Fujian Agriculture and Forestry University, China.
| | - Hua-Qin He
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
41
|
Bisht S, Singh MF. The triggering pathway, the metabolic amplifying pathway, and cellular transduction in regulation of glucose-dependent biphasic insulin secretion. Arch Physiol Biochem 2024; 130:854-865. [PMID: 38196246 DOI: 10.1080/13813455.2023.2299920] [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: 08/07/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Insulin secretion is a highly regulated process critical for maintaining glucose homeostasis. This abstract explores the intricate interplay between three essential pathways: The Triggering Pathway, The Metabolic Amplifying Pathway, and Cellular Transduction, in orchestrating glucose-dependent biphasic insulin secretion. MECHANISM During the triggering pathway, glucose metabolism in pancreatic beta-cells leads to ATP production, closing ATP-sensitive potassium channels and initiating insulin exocytosis. The metabolic amplifying pathway enhances insulin secretion via key metabolites like NADH and glutamate, enhancing calcium influx and insulin granule exocytosis. Additionally, the cellular transduction pathway involves G-protein coupled receptors and cyclic AMP, modulating insulin secretion. RESULT AND CONCLUSION These interconnected pathways ensure a dynamic insulin response to fluctuating glucose levels, with the initial rapid phase and the subsequent sustained phase. Understanding these pathways' complexities provides crucial insights into insulin dysregulation in diabetes and highlights potential therapeutic targets to restore glucose-dependent insulin secretion.
Collapse
Affiliation(s)
- Shradha Bisht
- Amity Institute of Pharmacy, Amity University, Lucknow, Uttar Pradesh, India
| | - Mamta F Singh
- School of Pharmaceutical Sciences, SBS University, Balawala, Uttarakhand, India
| |
Collapse
|
42
|
Ahmed I, Chakraborty R, Faizy AF, Moin S. Exploring the key role of DNA methylation as an epigenetic modulator in oxidative stress related islet cell injury in patients with type 2 diabetes mellitus: a review. J Diabetes Metab Disord 2024; 23:1699-1718. [PMID: 39610516 PMCID: PMC11599646 DOI: 10.1007/s40200-024-01496-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/21/2024] [Indexed: 11/30/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial metabolic disorder characterised by impaired insulin secretion and action, often exacerbated by oxidative stress. Recent research has highlighted the intricate involvement of epigenetic mechanisms, particularly DNA methylation, in the pathogenesis of T2DM. This review aims to elucidate the role of DNA methylation as an epigenetic modifier in oxidative stress-mediated beta cell dysfunction, a key component of T2DM pathophysiology. Oxidative stress, arising from an imbalance between reactive oxygen species (ROS) production and antioxidant defence mechanisms, is a hallmark feature of T2DM. Beta cells, responsible for insulin secretion, are particularly vulnerable to oxidative damage due to their low antioxidant capacity. Emerging evidence suggests that oxidative stress can induce aberrant DNA methylation patterns in beta cells, leading to altered gene expression profiles associated with insulin secretion and cell survival. Furthermore, studies have identified specific genes involved in beta cell function and survival that undergo DNA methylation changes in response to oxidative stress in T2DM. These epigenetic modifications can perpetuate beta cell dysfunction by dysregulating key pathways essential for insulin secretion, such as the insulin signalling cascade and mitochondrial function. Understanding the interplay between DNA methylation, oxidative stress, and beta cell dysfunction holds promise for developing novel therapeutic strategies for T2DM. Targeting aberrant DNA methylation patterns may offer new avenues for restoring beta cell function and improving glycemic control in patients with T2DM. However, further research is needed to elucidate the complex mechanisms underlying epigenetic regulation in T2DM and to translate these findings into clinical interventions.
Collapse
Affiliation(s)
- Istiaque Ahmed
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Ritoja Chakraborty
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Abul Faiz Faizy
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Shagufta Moin
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| |
Collapse
|
43
|
Prochaska M, Adeola G, Vetter N, Mirmira RG, Coe F, Worcester E. Insulin Resistance in Hypercalciuric Calcium Kidney Stone Patients. Kidney Med 2024; 6:100922. [PMID: 39634328 PMCID: PMC11615144 DOI: 10.1016/j.xkme.2024.100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Rational & Objective Diabetes and uric acid kidney stones are strongly associated. Patients with calcium kidney stones also have higher risk of developing diabetes compared with nonkidney stone patients yet this has not been further investigated. We aimed to characterize insulin resistance in calcium kidney stone patients. Study Design Observational. Setting & Population This study was performed in the University of Chicago Clinical Research Center. Kidney stone patients (N = 42) were selected for having idiopathic hypercalciuria and calcium stones with no other medical conditions, and controls (N = 27) were healthy. Exposures All participants presented to the Clinical Research Center in a fasting state and at least 2 timed fasting blood and urine collections were collected before a fixed breakfast. Six additional timed blood and urine collections were performed after breakfast. Outcomes We compared fasting and fed indices of insulin resistance between the groups. Analytic Approach We used t tests and multivariable linear regression models. A sensitivity analysis removing all patients who had ever been on a thiazide diuretic was also performed. Results In separate multivariable linear models, kidney stone patients had higher fasting serum insulin levels (24 (3-46 pmol/L), P = 0.03) and higher homeostatic model of insulin resistance (HOMA-IR) (1.0 (0.2-1.8), P = 0.02). In separate multivariable linear models, kidney stone patients had higher fed serum glucose levels (10 (2-18 mg/dL), P = 0.01). Results were similar in a sensitivity analysis removing all patients who had ever been on a thiazide diuretic. There were no differences in urine composition based on HOMA-IR levels. Limitations Single institution. Small sample size limited subanalyses by different calcium stone types. Conclusions Calcium kidney stone patients without diabetes or other medical conditions demonstrated signs of insulin resistance compared with healthy matched controls.
Collapse
Affiliation(s)
| | - Gloria Adeola
- Department of Medicine, University of Chicago, Chicago IL
| | - Noah Vetter
- Department of Medicine, University of Chicago, Chicago IL
| | | | - Fredric Coe
- Department of Medicine, University of Chicago, Chicago IL
| | | |
Collapse
|
44
|
Azari H, George M, Albracht-Schulte K. Gut Microbiota-microRNA Interactions and Obesity Pathophysiology: A Systematic Review of Integrated Studies. Int J Mol Sci 2024; 25:12836. [PMID: 39684547 DOI: 10.3390/ijms252312836] [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: 10/31/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is the fifth leading cause of death globally and its comorbidities put a high burden on societies and cause disability. In this review, we aim to summarize the interactions and crosstalk between gut microbiota and micro-RNA (miRNA) in obesity. We searched for the relevant literature through PubMed, Web of Science, Scopus, and Science Direct. The study design is registered in the international prospective register of systematic reviews (Prospero). According to the inclusion criteria, eight studies were eligible for assessment (two studies including human subjects and six studies including animal subjects). We report that the interactions of miRNA and gut microbiota in the context of obesity are diverse and in some cases tissue specific. However, the interactions mediate obesity-associated pathways including the inflammatory response, oxidative stress, insulin signaling, gut permeability, and lipogenesis. To mention the most meaningful results, the expression of adipose tissue miRNA-378a-3p/5p was associated with Bifidobacterium and Akkermansia abundance, the expression of hepatic miRNA-34a was related to the Firmicutes phylum, and the expression of miRNA-122-5p and miRNA-375 was associated with the Bacteroides genus. miRNA-microbiota-associated pathological pathways seem to provide an intricate, but promising field for future research directed toward the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Hushyar Azari
- Department of Kinesiology and Sport Management and Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Megan George
- Department of Kinesiology and Sport Management and Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Kembra Albracht-Schulte
- Department of Kinesiology and Sport Management and Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
45
|
Jacques E, van den Bosch A, de Vos van Steenwijk P, Kooreman L, Delvoux B, Romano A, Werner H. Pilot Data Suggest That Obesity and Presence of Malignancy Are Associated with Altered Immune Cell Infiltration in Endometrial Biopsies. J Clin Med 2024; 13:7248. [PMID: 39685707 DOI: 10.3390/jcm13237248] [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: 09/25/2024] [Revised: 11/01/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
(1) Background: The worldwide endometrial cancer (EC) incidence is rising, amongst others linked to obesity, type 2 diabetes mellitus (T2DM), and metabolic syndrome, possibly due to low-grade adipose tissue inflammation. We studied immune cell infiltration in the endometrium in relation to diagnosis and obesity. (2) Methods: A cohort was created (n = 44) from postmenopausal women, lean (n = 15) and obese (n = 29), with bleeding complaints due to EC (n = 18) or benign pathology (n = 26). Endometrial biopsies were used to study the immune microenvironment and stained for macrophages (CD68 and CD163), T-cells (CD3 and CD8), and NK-cells (CD56). (3) Results: Malignant samples showed reduced intraepithelial CD3+ and CD8+ T-cells and increased stromal CD3+ T-cells. In obese patients, increased intraepithelial CD3+ and CD8+ T-cells were detected, especially in obese patients with T2DM. Epithelial CD56+ NK-cells were depleted in EC; however, no effect of obesity on NK-cell infiltration was observed. Stromal CD68+ cells were reduced in EC patients, whereas the CD163+ cells were increased. (4) Conclusions: Obesity and malignancy are associated with differences in immune cell presence. The alterations in immune cell infiltration seen in obese EC patients with and without diabetes suggest a complex interaction where obesity-related low-grade inflammation plays a central role.
Collapse
Affiliation(s)
- Eline Jacques
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Anouk van den Bosch
- GROW-Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Peggy de Vos van Steenwijk
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
- GROW-Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Loes Kooreman
- GROW-Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Bert Delvoux
- GROW-Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Andrea Romano
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
- GROW-Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Henrica Werner
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
- GROW-Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
46
|
Cao W, Liu Y, Wei H, Dong Y, Sun H, Zhang X, Qiu J. Aerobic exercise attenuates insulin resistance via restoring branched chain amino acids homeostasis in obese mice. Front Nutr 2024; 11:1451429. [PMID: 39634544 PMCID: PMC11615396 DOI: 10.3389/fnut.2024.1451429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Emerging evidences suggests that the disrupted branched-chain amino acids (BCAAs) homeostasis and elevated BCAAs promote obesity-related insulin resistance (IR). Exercise improves insulin sensitivity. However, whether BCAAs plays a role in the exercise-attenuated IR remains to be fully investigated. Methods In this study, male C57BL/6J mice were induced to become diet-induced obese (DIO) and served as subjects. The initial investigation focused on the impact of exercise on IR and BCAAs. The DIO mice were randomly assigned to either a sedentary group (CON, n = 16) or an exercise group (EX, n = 16). The EX group underwent a 12-week aerobic exercise regimen on a treadmill. After 12-week, plasma BCAAs and branched-chain keto acids (BCKAs) were measured by liquid chromatography-mass spectrometry, glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed, and the expression and phosphorylation of BCAAs catabolic proteins, as well as AKT T308 in gastrocnemius muscle and liver tissues, were evaluated using western blotting. Subsequently, the study explored the role of BCAAs in enhancing IR through exercise. Mice were randomly allocated into 4 groups: sedentary group (CON, n = 8), sedentary with BCAAs supplementation group (CON+BCAA, n = 8), exercise group (EX, n = 16), and exercise with BCAAs supplementation group (EX+BCAA, n = 16). The exercise protocol was as above. Mice in the BCAAs supplemented groups received drinking water containing 2% BCAAs. After 12-week, plasma BCAAs and BCKAs were measured, GTT and ITT tests were performed, and the phosphorylation of AKT T308, as well as p70S6K T389 in gastrocnemius muscle and liver, were compared between the EX group and the EX+BCAA group. Additionally, the phosphorylation of AMPKα T172 in both tissues was measured across all four groups. Results 12-week aerobic exercise improved insulin sensitivity in DIO mice while inducing BCAAs catabolic protein expression in skeletal muscle and liver, and reducing the plasma BCAAs level. Importantly, BCAAs supplementation elevated the plasma level of BCAAs and counteracted the exercise-attenuated IR. In skeletal muscle and liver tissues, BCAAs supplementation impaired the exercise-improved insulin signaling without enhancing mammalian target of rapamycin activity. AMPK activity was enhanced by aerobic exercise, which was abolished by BCAAs supplementation. Conclusion Aerobic exercise attenuated insulin resistance via restoring BCAAs homeostasis and AMPK activity. The impacts of BCAAs intake on the metabolic effects of exercise sheds light on the combined exercise and nutrition intervention strategy for diabetes management.
Collapse
Affiliation(s)
- Wei Cao
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
- College of Sports and Health, Shandong Sport University, Rizhao, China
| | - Yajin Liu
- Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Hao Wei
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Yunfeng Dong
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Haipeng Sun
- Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Center for Cardiovascular Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Xuejiao Zhang
- Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Center for Cardiovascular Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Junqiang Qiu
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
- Beijing Sports Nutrition Engineering Research Center, Beijing, China
| |
Collapse
|
47
|
Huo M, Yu X, Yuan X, Guo J, Wei B, Shi Y, Gu Y, Zhang X, Sun M. The P300-ARRDC3 axis participates in maternal subclinical hypothyroidism and is involved in abnormal hepatic insulin sensitivity in adult offspring. Heliyon 2024; 10:e39259. [PMID: 39568856 PMCID: PMC11577204 DOI: 10.1016/j.heliyon.2024.e39259] [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: 02/18/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Numerous studies have suggested potential associations between maternal subclinical hypothyroidism (SCH) and adverse metabolic outcomes in offspring, however, the underlying mechanism remains unclear. In this study, we generated a maternal SCH mouse model by administering 50 ppm 6-propyl-2-thiouracil (PTU) in the drinking water of pregnant mice until delivery. This model was used to investigate the mechanisms influencing glucose metabolism in offspring. RNA sequencing (RNA-seq) revealed a substantial increase in ARRDC3 expression in the livers of the offspring of the SCH model mice, which may contribute to insulin resistance. Additionally, the phosphorylation levels of key proteins in the insulin signalling pathway, such as protein kinase B (Akt), glycogen synthase kinase 3 beta (GSK-3β), and Forkhead box protein O1 (FoxO1), were correspondingly reduced in the SCH offspring. Moreover, overexpression of ARRDC3 in Hepa1‒6 cells suppressed the Akt/GSK-3β/FoxO1 signalling pathway and increased the expression of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), which was consistent with the molecular changes observed in SCH offspring. Our results also indicated that the upregulation of ARRDC3 in SCH offspring may result from increased H3K27 acetylation of the ARRDC3 promoter region, driven by elevated expression of P300. Importantly, adequate L-T4 supplementation during pregnancy improved insulin sensitivity and reversed the molecular alterations in the insulin signalling pathway observed in SCH offspring. In conclusion, exposure to intrauterine SCH resulted in altering the P300-ARRDC3 axis in offspring and impaired insulin sensitivity by disrupting the Akt/GSK-3β/FoxO1 signalling pathway. Timely L-T4 supplementation during pregnancy is an effective strategy to prevent insulin resistance in offspring of SCH mothers. This study elucidates potential molecular mechanisms behind insulin resistance in SCH offspring and suggests novel therapeutic targets for treating metabolic disorders related to maternal SCH.
Collapse
Affiliation(s)
- Ming Huo
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou City, 730000, Gansu, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xianbin Yuan
- The first people's hospital of Lanzhou City, Lanzhou City, 730000, Gansu, China
| | - Jun Guo
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yannan Gu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xuehong Zhang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou City, 730000, Gansu, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
48
|
Qiu J, He S, Yu C, Yang R, Kuang M, Sheng G, Zou Y. Assessing the validity of METS-IR for predicting the future onset of diabetes: an analysis using time-dependent receiver operating characteristics. BMC Endocr Disord 2024; 24:238. [PMID: 39508243 PMCID: PMC11542444 DOI: 10.1186/s12902-024-01769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The Metabolic Insulin Resistance Score (METS-IR) is a non-invasive proxy for insulin resistance (IR) that has been newly developed in recent years and has been shown to be associated with diabetes risk. Our aim was to assess the predictive value of METS-IR for the future development of diabetes and its temporal differences in people of different sex, age, and body mass index (BMI). METHODS The current study included 15,453 baseline non-diabetic subjects in the NAGALA cohort and then grouped according to the World Health Organization's (WHO) recommended criteria for age and BMI. Multivariate Cox regression and time-dependent receiver operator characteristics (ROC) curves were used to analyze the value of METS-IR in assessing and predicting the risk of diabetes in people of different sexes, ages, and BMIs. RESULTS 373 individuals developed diabetes during the observation period. By multivariate COX regression analysis, the development of future diabetes was significantly associated with increased METS-IR, and this positive association was stronger in women than in men and in individuals < 45 years than in individuals ≥ 45 years; while no significant differences were observed between non-obese and overweight/obesity individuals. Using time-dependent ROC analysis we also assessed the predictive value of METS-IR for future diabetes at a total of 11-time points between 2 and 12 years. The results showed that METS-IR had a higher predictive value for the future development of diabetes in women or individuals < 45 years of age compared to men or individuals ≥ 45 years of age for almost the entire follow-up period. Furthermore, across different BMI categories, we also found that in the short term (3-5 years), METS-IR had a higher predictive value for the development of diabetes in individuals with overweight/obesity, while in the medium to long term (6-12 years), METS-IR was more accurate in predicting the development of diabetes in non-obese individuals. CONCLUSIONS Our study showed that METS-IR was independently associated with the development of future diabetes in a non-diabetic population. METS-IR was a good predictor of diabetes, especially for women and individuals < 45 years old for predicting the future risk of developing diabetes at all times.
Collapse
Affiliation(s)
- Jiajun Qiu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shiming He
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Provincial, China
| | - Changhui Yu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Provincial, China
| | - Ruijuan Yang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Endocrinology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Provincial, China
| | - Maobin Kuang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Provincial, China
| | - Guotai Sheng
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Provincial, China.
| | - Yang Zou
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Provincial, China.
| |
Collapse
|
49
|
Miao X, Alidadipour A, Saed V, Sayyadi F, Jadidi Y, Davoudi M, Amraee F, Jadidi N, Afrisham R. Hepatokines: unveiling the molecular and cellular mechanisms connecting hepatic tissue to insulin resistance and inflammation. Acta Diabetol 2024; 61:1339-1361. [PMID: 39031190 DOI: 10.1007/s00592-024-02335-9] [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: 05/04/2024] [Accepted: 07/06/2024] [Indexed: 07/22/2024]
Abstract
Insulin resistance arising from Non-Alcoholic Fatty Liver Disease (NAFLD) stands as a prevalent global ailment, a manifestation within societies stemming from individuals' suboptimal dietary habits and lifestyles. This form of insulin resistance emerges as a pivotal factor in the development of type 2 diabetes mellitus (T2DM). Emerging evidence underscores the significant role of hepatokines, as hepatic-secreted hormone-like entities, in the genesis of insulin resistance and eventual onset of type 2 diabetes. Hepatokines exert influence over extrahepatic metabolism regulation. Their principal functions encompass impacting adipocytes, pancreatic cells, muscles, and the brain, thereby playing a crucial role in shaping body metabolism through signaling to target tissues. This review explores the most important hepatokines, each with distinct influences. Our review shows that Fetuin-A promotes lipid-induced insulin resistance by acting as an endogenous ligand for Toll-like receptor 4 (TLR-4). FGF21 reduces inflammation in diabetes by blocking the nuclear translocation of nuclear factor-κB (NF-κB) in adipocytes and adipose tissue, while also improving glucose metabolism. ANGPTL6 enhances AMPK and insulin signaling in muscle, and suppresses gluconeogenesis. Follistatin can influence insulin resistance and inflammation by interacting with members of the TGF-β family. Adropin show a positive correlation with phosphoenolpyruvate carboxykinase 1 (PCK1), a key regulator of gluconeogenesis. This article delves into hepatokines' impact on NAFLD, inflammation, and T2DM, with a specific focus on insulin resistance. The aim is to comprehend the influence of these recently identified hormones on disease development and their underlying physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Xiaolei Miao
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Arian Alidadipour
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Vian Saed
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Firooze Sayyadi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Jadidi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Davoudi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amraee
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Jadidi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Eid SA, Elzinga SE, Kim B, Rumora AE, Hayes JM, Carter A, Pacut C, Allouch AM, Koubek EJ, Feldman EL. High-Intensity Interval Training, Caloric Restriction, or Their Combination Have Beneficial Effects on Metabolically Acquired Peripheral Neuropathy. Diabetes 2024; 73:1895-1907. [PMID: 39163551 PMCID: PMC11493763 DOI: 10.2337/db23-0997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Peripheral neuropathy (PN) is a prevalent and debilitating complication of obesity, prediabetes, and type 2 diabetes, which remains poorly understood and lacks disease-modifying therapies. Fortunately, diet and/or exercise have emerged as effective treatment strategies for PN. Here, we examined the impact of caloric restriction (CR) and high-intensity interval training (HIIT) interventions, alone or combined (HIIT-CR), on metabolic and PN outcomes in high-fat diet (HFD) mice. HFD feeding alone resulted in obesity, impaired glucose tolerance, and PN. Peripheral nerves isolated from these mice also developed insulin resistance (IR). CR and HIIT-CR, but not HIIT alone, improved HFD-induced metabolic dysfunction. However, all interventions improved PN to similar extents. When examining the underlying neuroprotective mechanisms in whole nerves, we found that CR and HIIT-CR activate the fuel-sensing enzyme AMPK. We then performed complimentary in vitro work in Schwann cells, the glia of peripheral nerves. Treating primary Schwann cells with the saturated fatty acid palmitate to mimic prediabetic conditions caused IR, which was reversed by the AMPK activator, AICAR. Together, these results enhance our understanding of PN pathogenesis, the differential mechanisms by which diet and exercise may improve PN, and Schwann cell-specific contributions to nerve insulin signaling and PN progression. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Amy E. Rumora
- Department of Neurology, University of Michigan, Ann Arbor, MI
- Department of Neurology, Columbia University, New York, NY
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Andrew Carter
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Adam M. Allouch
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI
| |
Collapse
|