1
|
Fojas EG, Haidery A, Naseeb S, Naemi R. Type 2 diabetes and cardiovascular conditions prediction in individuals with metabolic syndrome-associated lipoprotein lipase gene (LPL) single nucleotide polymorphisms (SNPs). J Diabetes Complications 2025; 39:109003. [PMID: 40179477 DOI: 10.1016/j.jdiacomp.2025.109003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE Metabolic syndrome (MetS) is predictive of increased risk of type 2 diabetes (T2D) and cardiovascular conditions (CVC). Lipoprotein lipase gene (LPL) single nucleotide polymorphisms (SNPs) may be of importance to the eventual diagnosis of T2D and CVC. This study aimed to predict the diagnosis of T2D and CVC amongst individuals with LPL SNPs rs268, rs11542065, rs116403115, rs118204057, rs118204061, rs144466625, and rs547644955. METHODS This is a retrospective study using the UK Biobank data. Variables associated with MetS, T2D and CVC were selected from the data set. The total number of subjects in the cohort was 12,872 (mean age 56 years ± 8.1, 90.0 % were of British ethnicity, and 53.9 % were females). Logistic regression was used to assess whether the T2D and CVC can be predicted based on the presence of LPL SNPs and some of the clinical measures. RESULTS Prediction models using clinical parameters showed good area under the curve (AUC) for prediction of T2D and CVC diagnosis (in receiver operating characteristic (ROC) analysis, area under the curve (AUC) = 0.959 for T2D, AUC = 0.772 for CVC). The addition of Polygenic Risk Scores (PRS/s) showed an improvement for diagnosis of both (AUC = 0.961 and 0.790 for TD and CVC, respectively). Further addition of SNPs showed more increase in AUC (AUC = 0.965 and 0.837 for T2D and CVC, respectively). The additive effect of the PRSs and LPL SNPs was more pronounced in the CVC than in the T2D model. The variant that had major significance for both T2D and CVC diagnoses was rs547644955 (AUC 1.0 and 0.910, respectively). The SNPs rs116403115 and rs118204057 both had an AUC of 1.0 for T2D diagnosis. CONCLUSION The prediction of T2D and CVC diagnoses with the use of clinically available factors may be enhanced with the addition of PRSs and SNPs, including LPL SNPs, which may have implications for stratified or personalised approaches for disease prevention or treatment.
Collapse
Affiliation(s)
- Esphie Grace Fojas
- University of Staffordshire, School of Health, Education, Policing and Sciences, Stoke-on-Trent, England, United Kingdom.
| | - Ahmad Haidery
- University of Staffordshire, School of Health, Education, Policing and Sciences, Stoke-on-Trent, England, United Kingdom
| | - Samina Naseeb
- The University of Manchester, School of Biological Sciences, Faculty of Biology Medicine and Health, England, United Kingdom
| | - Roozbeh Naemi
- University of Salford, School of Health and Society, Manchester, England, United Kingdom
| |
Collapse
|
2
|
Mir MM, Alghamdi M, BinAfif WF, Alharthi MH, Alshahrani AM, Alamri MMS, Alfaifi J, Ameer AYA, Mir R. Emerging biomarkers in type 2 diabetes mellitus. Adv Clin Chem 2025; 126:155-198. [PMID: 40185534 DOI: 10.1016/bs.acc.2025.01.002] [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: 04/07/2025]
Abstract
Diabetes mellitus is a chronic condition caused by high blood glucose resulting from insufficient insulin production or cellular resistance to insulin action or both. It is one of the fastest-growing public health concerns worldwide. Development of long-term nephropathy, retinopathy, neuropathy, and cardiovascular disease are some of the complications commonly associated with poor blood glycemic control. Type 2 diabetes mellitus (T2DM), the most prevalent type of diabetes, accounts for around 95 % of all cases globally. Although middle-aged or older adults are more likely to develop T2DM, its prevalence has grown in children and young people due to increased obesity, sedentary lifestyle and poor nutrition. Furthermore, it is believed that more than 50 % of cases go undiagnosed annually. Routine screening is essential to ensure early detection and reduce risk of life-threatening complications. Herein, we review traditional biomarkers and highlight the ongoing pursuit of novel and efficacious biomarkers driven by the objective of achieving early, precise and prompt diagnoses. It is widely acknowledged that individual biomarkers will inevitably have certain limitations necessitating the need for integrating multiple markers in screening.
Collapse
Affiliation(s)
- Mohammad Muzaffar Mir
- Departments of Clinical Biochemistry, College of Medicine, University of Bisha, Bisha, Saudi Arabia.
| | - Mushabab Alghamdi
- Internal Medicine, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Waad Fuad BinAfif
- Internal Medicine, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Muffarah Hamid Alharthi
- Family and Community Medicine, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Abdullah M Alshahrani
- Family and Community Medicine, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | | | - Jaber Alfaifi
- Child Health, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | | | - Rashid Mir
- Prince Fahd Bin Sultan Research Chair, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
3
|
Han M, Shen Y, Guo X, Hong C, Ji X, Guo H, Jin Y, Yuan H. Association between non-high-density lipoprotein cholesterol and type 2 diabetes: a systematic review and meta-analysis of cohort studies. Endocr J 2025; 72:43-51. [PMID: 39313371 PMCID: PMC11778345 DOI: 10.1507/endocrj.ej24-0189] [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: 04/02/2024] [Accepted: 08/24/2024] [Indexed: 09/25/2024] Open
Abstract
Non-high-density lipoprotein cholesterol (non-HDL), a more readily available and reliable lipid parameter, is unclear in its association with type 2 diabetes (T2D). Previous studies assessing the relationship between non-HDL and T2D risk remains inconsistent results. We performed a meta-analysis to systematically evaluate this association. The PubMed, EMBASE, Medline, Web of Science, and Cochrane Library databases were systematically searched to find articles on "non-HDL" and "T2D" from inception to December 6, 2023. A random-effects model was used to calculate the effect estimates and 95% confidence intervals. Subgroup analyses and univariate Meta-regression were performed to explore sources of heterogeneity. The main exposure and outcome were non-HDL and T2D, respectively, in the general population. A total of 8 studies included 251,672 participants who met the inclusion criteria for this study. Meta-analysis showed that higher non-HDL increased the risk of T2D compared with the lower non-HDL group (total effect size: 1.16; 95% CI 1.079-1.251, p < 0.001). Subgroup analyses and Meta-regression of the association between non-HDL and T2D were not affected by region, proportion of men, sample size, or adjustment for confounders (including BMI, hypertension, waist circumference, and family history of diabetes). Higher non-HDL may be associated with an increased risk of T2D. Large prospective cohort studies are needed to validate these findings, and further studies are required in order to elucidate the underlying pathophysiologic mechanisms underlying the association between non-HDL and T2D.
Collapse
Affiliation(s)
- Mengqi Han
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Yue Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Xin Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Cheng Hong
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Xincan Ji
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Haoyang Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Yuelong Jin
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Hui Yuan
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu 241002, China
| |
Collapse
|
4
|
Codazzi V, Frontino G, Galimberti L, Giustina A, Petrelli A. Mechanisms and risk factors of metabolic syndrome in children and adolescents. Endocrine 2024; 84:16-28. [PMID: 38133765 PMCID: PMC10987369 DOI: 10.1007/s12020-023-03642-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS) is a complex disorder characterized by abdominal obesity, elevated blood pressure, hyperlipidemia, and elevated fasting blood glucose levels. The diagnostic criteria for MetS in adults are well-established, but there is currently no consensus on the definition in children and adolescents. The etiology of MetS is believed to involve a complex interplay between genetic predisposition and environmental factors. While genetic predisposition explains only a small part of MetS pathogenesis, modifiable environmental risk factors play a significant role. Factors such as maternal weight during pregnancy, children's lifestyle, sedentariness, high-fat diet, fructose and branched-chain amino acid consumption, vitamin D deficiency, and sleep disturbances contribute to the development of MetS. Early identification and treatment of MetS in children and adolescents is crucial to prevent the development of chronic diseases later in life. In this review we discuss the latest research on factors contributing to the pathogenesis of MetS in children, focusing on non-modifiable and modifiable risk factors, including genetics, dysbiosis and chronic low-grade inflammation.
Collapse
|
5
|
Flori L, Piragine E, Calderone V, Testai L. Role of hydrogen sulfide in the regulation of lipid metabolism: Implications on cardiovascular health. Life Sci 2024; 341:122491. [PMID: 38336275 DOI: 10.1016/j.lfs.2024.122491] [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: 12/06/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The World Health Organization (WHO) defines obesity as an urgency for health and a social emergency. Today around 39 % of people is overweight, of these over 13 % is obese. It is well-consolidated that the adipose cells are deputy to lipid storage under caloric excess; however, despite the classical idea that adipose tissue has exclusively a passive function, now it is known to be deeply involved in the regulation of systemic metabolism in physiological as well as under obesogenic conditions, with consequences on cardiovascular health. Beside two traditional types of adipose cells (white and brown), recently the beige one has been highlighted as the consequence of the healthy remodeling of white adipocytes, confirming their metabolic adaptability. In this direction, pharmacological, nutraceutical and nutrient-based approaches are addressed to positively influence inflammation and metabolism, thus contributing to reduce the obese-associated cardiovascular risk. In this scenario, hydrogen sulfide emerges as a new mediator that may regulate crucial targets involved in the regulation of metabolism. The current evidence demonstrates that hydrogen sulfide may induce peroxisome proliferator activated receptor γ (PPARγ), a crucial mediator of adipogenesis, inhibit the phosphorylation of perlipin-1 (plin-1), a protein implicated in the lipolysis, and finally promote browning process, through the release of irisin from skeletal muscle. The results summarized in this review suggest an important role of hydrogen sulfide in the regulation of metabolism and in the prevention/treatment of obese-associated cardiovascular diseases and propose new insight on the putative mechanisms underlying the release of hydrogen sulfide or its biosynthesis, delineating a further exciting field of application.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Eugenia Piragine
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Lara Testai
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| |
Collapse
|
6
|
Yuge H, Okada H, Hamaguchi M, Kurogi K, Murata H, Ito M, Fukui M. Triglycerides/HDL cholesterol ratio and type 2 diabetes incidence: Panasonic Cohort Study 10. Cardiovasc Diabetol 2023; 22:308. [PMID: 37940952 PMCID: PMC10634002 DOI: 10.1186/s12933-023-02046-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Previous studies have investigated the association between the ratio of triglycerides (TG) to high-density lipoprotein cholesterol (HDL-C) and the incidence of diabetes in adults and discovered that a high TG/HDL-C ratio was linked to an elevated risk of new-onset diabetes. However, the comparison of predicting diabetes development among lipid profiles including the TG/HDL-C ratio, and the ratio of TG/HDL-C cut-off value has received limited attention. We examined the relationship between diabetes onset and the TG/HDL-C ratio in addition to the applicable cut-off value for predicting diabetes onset. METHODS This study included 120,613 participants from the health examination database at Panasonic Corporation from 2008 to 2017. Cox regression analysis employing multivariable models was used to investigate the association between lipid profiles, particularly the ratio of TG/HDL-C and the development of type 2 diabetes (T2D). The multivariable model was adjusted for age, sex, BMI, systolic blood pressure, plasma glucose levels after fasting, smoking status, and exercise habits. Areas under time-dependent receiver operating characteristic (ROC) curves (AUCs) were employed to assess the prediction performance and cut-off values of each indicator. A fasting plasma glucose level of 126 mg/dL, a self-reported history of diabetes, or usage of antidiabetic medicines were used to identify T2D. RESULTS During the course of the study, 6,080 people developed T2D. The median follow-up duration was 6.0 (3-10) years. Multivariable analysis revealed that the ratio of TG/HDL-C (per unit, HR; 1.03 [95% CI 1.02-1.03]) was substantially linked to the risk of incident T2D. AUC and cut-off points for the ratio of TG/HDL-C for T2D development after 10 years were 0.679 and 2.1, respectively. Furthermore, the AUC of the ratio of TG/HDL-C was considerably larger compared to that of LDL-C, HDL-C, and TG alone (all P < 0.001). We discovered an interaction effect between sex, BMI, and lipid profiles in subgroup analysis. Females and participants having a BMI of < 25 kg/m2 showed a higher correlation between lipid profile levels and T2D onset. CONCLUSIONS The ratio of TG/HDL-C was found to be a stronger predictor of T2D development within 10 years than LDL-C, HDL-C, or TG, indicating that it may be useful in future medical treatment support.
Collapse
Affiliation(s)
- Hiroki Yuge
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
- Department of Diabetes and Endocrinology, Matsushita Memorial Hospital, 5-55 Sotojima-Cho, Moriguchi, 570-8540, Japan.
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Kazushiro Kurogi
- Department of Health Care Center, Panasonic Health Insurance Organization, 5-55 Sotojima-Cho, Moriguchi, 570-8540, Japan
| | - Hiroaki Murata
- Department of Orthopedic Surgery, Matsushita Memorial Hospital, 5-55 Sotojima-Cho, Moriguchi, 570-8540, Japan
| | - Masato Ito
- Department of Health Care Center, Panasonic Health Insurance Organization, 5-55 Sotojima-Cho, Moriguchi, 570-8540, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| |
Collapse
|
7
|
Zhao YK, Zhu XD, Liu R, Yang X, Liang YL, Wang Y. The Role of PPARγ Gene Polymorphisms, Gut Microbiota in Type 2 Diabetes: Current Progress and Future Prospects. Diabetes Metab Syndr Obes 2023; 16:3557-3566. [PMID: 37954888 PMCID: PMC10638901 DOI: 10.2147/dmso.s429825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
Over the past decade, there has been a significant increase in studies investigating the relationship between the polymorphisms of the Peroxisome Proliferator-Activated Receptor gamma (PPARγ) gene and Type 2 Diabetes (T2D). PPARγ, a critical transcription factor, plays a central role in lipid metabolism, insulin resistance, and inflammatory response. Concurrently, the influence of gut microbiota on the development of T2D has gained increasing attention, especially their role in affecting host metabolism, such as lipid metabolism and the PPARγ signaling pathway. This review provides a comprehensive analysis of recent studies on PPARγ gene polymorphisms and their association with T2D, with a specific emphasis on the implications of gut microbiota and their interaction with PPARγ pathways. We also discuss the potential of manipulating gut microbiota and targeting PPARγ gene polymorphisms in T2D management. By deepening our understanding of these relationships, we aim to pave the way for novel preventative and therapeutic strategies for T2D.
Collapse
Affiliation(s)
- Yi-Kun Zhao
- Department of Basic Medical College, Gansu University of Chinese Medicine, Lanzhou City, People’s Republic of China
| | - Xiang-Dong Zhu
- Department of Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan city, People’s Republic of China
| | - Rong Liu
- Department of Basic Medical College, Gansu University of Chinese Medicine, Lanzhou City, People’s Republic of China
| | - Xia Yang
- Department of Basic Medical College, Gansu University of Chinese Medicine, Lanzhou City, People’s Republic of China
| | - Yong-Lin Liang
- Department of Basic Medical College, Gansu University of Chinese Medicine, Lanzhou City, People’s Republic of China
| | - Yan Wang
- Department of Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan city, People’s Republic of China
| |
Collapse
|
8
|
Preda A, Carbone F, Tirandi A, Montecucco F, Liberale L. Obesity phenotypes and cardiovascular risk: From pathophysiology to clinical management. Rev Endocr Metab Disord 2023; 24:901-919. [PMID: 37358728 PMCID: PMC10492705 DOI: 10.1007/s11154-023-09813-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 06/27/2023]
Abstract
Obesity epidemic reached the dimensions of a real global health crisis with more than one billion people worldwide living with obesity. Multiple obesity-related mechanisms cause structural, functional, humoral, and hemodynamic alterations with cardiovascular (CV) deleterious effects. A correct assessment of the cardiovascular risk in people with obesity is critical for reducing mortality and preserving quality of life. The correct identification of the obesity status remains difficult as recent evidence suggest that different phenotypes of obesity exist, each one associated with different degrees of CV risk. Diagnosis of obesity cannot depend only on anthropometric parameters but should include a precise assessment of the metabolic status. Recently, the World Heart Federation and World Obesity Federation provided an action plan for management of obesity-related CV risk and mortality, stressing for the instauration of comprehensive structured programs encompassing multidisciplinary teams. In this review we aim at providing an updated summary regarding the different obesity phenotypes, their specific effects on CV risk and differences in clinical management.
Collapse
Affiliation(s)
| | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Amedeo Tirandi
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy.
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy.
| | - Luca Liberale
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| |
Collapse
|
9
|
Hu LK, Liu YH, Yang K, Chen N, Ma LL, Yan YX. Association between hypertriglyceridemic-waist phenotype and circadian syndrome risk: a longitudinal cohort study. Hormones (Athens) 2023; 22:457-466. [PMID: 37423976 DOI: 10.1007/s42000-023-00462-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
Recently, circadian syndrome (CircS) has been proposed as a new predictor of cardiometabolic risk. We aimed to investigate the relationship between the hypertriglyceridemic-waist phenotype and its dynamic status with CircS in China. We conducted a two-stage study based on the China Health and Retirement Longitudinal Study (CHARLS) from 2011 to 2015. Multivariate logistic regression models in cross-sectional analysis and Cox proportional hazards regression models in longitudinal analysis were used to estimate the associations of hypertriglyceridemic-waist phenotypes with CircS and its components. We then applied multiple logistic regression analysis to evaluate the odds ratios (ORs) and 95% confidence intervals (CIs) for CircS risk by transformation into the hypertriglyceridemic-waist phenotype. A total of 9863 participants were included in the cross-sectional analysis and 3884 participants in the longitudinal analysis. Compared with normal waist circumference (WC) and normal triglyceride (TG) level (NWNT), CircS risk was increased with enlarged WC and high TG level (EWHT) (hazard ratio (HR) 3.87 [95% CI: 2.38, 5.39]). Similar results were observed in subgroup analyses by sex, age, smoking status, and drinking status. During follow-up, CircS risk was increased in group K (stable EWNT during follow-up) (OR 9.97 [95% CI: 6.41, 15.49]) compared with group A (stable NWNT during follow-up), while group L (baseline enlarged WC and normal TG level transformed to follow-up EWHT) had the highest risk of CircS (OR 116.07 [95% CI: 72.77, 185.14]). In conclusion, the hypertriglyceridemic-waist phenotype and its dynamic status were associated with the risk of developing CircS in Chinese adults.
Collapse
Affiliation(s)
- Li-Kun Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmenWai, Fengtai District, Beijing, 100069, China
| | - Yu-Hong Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmenWai, Fengtai District, Beijing, 100069, China
| | - Kun Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmenWai, Fengtai District, Beijing, 100069, China
| | - Ning Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmenWai, Fengtai District, Beijing, 100069, China
| | - Lin-Lin Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmenWai, Fengtai District, Beijing, 100069, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmenWai, Fengtai District, Beijing, 100069, China.
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, 100069, China.
| |
Collapse
|
10
|
Sekiya M, Ma Y, Kainoh K, Saito K, Yamazaki D, Tsuyuzaki T, Chen W, Adi Putri PIP, Ohno H, Miyamoto T, Takeuchi Y, Murayama Y, Sugano Y, Osaki Y, Iwasaki H, Yahagi N, Suzuki H, Motomura K, Matsuzaka T, Murata K, Mizuno S, Takahashi S, Shimano H. Loss of CtBP2 may be a mechanistic link between metabolic derangements and progressive impairment of pancreatic β cell function. Cell Rep 2023; 42:112914. [PMID: 37557182 DOI: 10.1016/j.celrep.2023.112914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/19/2023] [Accepted: 07/16/2023] [Indexed: 08/11/2023] Open
Abstract
The adaptive increase in insulin secretion in early stages of obesity serves as a safeguard mechanism to maintain glucose homeostasis that cannot be sustained, and the eventual decompensation of β cells is a key event in the pathogenesis of diabetes. Here we describe a crucial system orchestrated by a transcriptional cofactor CtBP2. In cultured β cells, insulin gene expression is coactivated by CtBP2. Global genomic mapping of CtBP2 binding sites identifies a key interaction between CtBP2 and NEUROD1 through which CtBP2 decompacts chromatin in the insulin gene promoter. CtBP2 expression is diminished in pancreatic islets in multiple mouse models of obesity, as well as human obesity. Pancreatic β cell-specific CtBP2-deficient mice manifest glucose intolerance with impaired insulin secretion. Our transcriptome analysis highlights an essential role of CtBP2 in the maintenance of β cell integrity. This system provides clues to the molecular basis in obesity and may be targetable to develop therapeutic approaches.
Collapse
Affiliation(s)
- Motohiro Sekiya
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan.
| | - Yang Ma
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Kenta Kainoh
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Kenji Saito
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Daichi Yamazaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Tomomi Tsuyuzaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Wanpei Chen
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Putu Indah Paramita Adi Putri
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Hiroshi Ohno
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Takafumi Miyamoto
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Yoshinori Takeuchi
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Yuki Murayama
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Yoko Sugano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Yoshinori Osaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Hitoshi Iwasaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Naoya Yahagi
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Hiroaki Suzuki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Kaori Motomura
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Takashi Matsuzaka
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan; Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Kazuya Murata
- Laboratory Animal Resource Center in Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| |
Collapse
|
11
|
Li K, Cao B, Ke J, Yang L, Zhao D. Association of Hyper-Triglyceridemic Waist Phenotype and Diabetic Vascular Complication in the Chinese Population. Diabetes Metab Syndr Obes 2023; 16:2233-2241. [PMID: 37525822 PMCID: PMC10387281 DOI: 10.2147/dmso.s416668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023] Open
Abstract
Background Diabetic vascular complications are the leading cause of crippling and death of diabetic patients and seriously affect patients' quality of life. It is essential to control the risk factors contributing to vascular complications in patients with Type 2 diabetes (T2DM). This study aimed to examine the association between hyper-triglyceridemic waist phenotype (HWP) and the risk of vascular complication index of diabetes in T2DM patients. Methods The participants with type 2 diabetes mellitus in this study registered at the National Metabolic Management Center (MMC) of Beijing Luhe Hospital from June 2017 to June 2021. Data were collected by trained personnel according to the protocol. The questionnaire containing information on demographic characteristics and lifestyle factors (including alcohol drinking and cigarette smoking et al) was administered by trained interviewers. Logistic regression analysis assessing the associations between the hyper-triglyceridemic waist phenotype and vascular complication index of diabetes. In addition, the subgroup analysis was performed by age, sex, HbA1c, hypertension or not, and education level. Results After data cleaning, a total of 3221 participants with T2DM were enrolled. The median (IQR) duration of diabetes was 47.00 (3.00, 125.00) months. Compared to the participants in the Normal triglycerides level and Normal waist circumference group (NTNW), those in the Elevated triglycerides level and Enlarged waist circumference group (HTGW) have a higher risk of CKD-related vascular complications; the OR of decreased estimated glomerular filtration rate (GFR) and elevated urinary albumin creatinine ratio (UACR) were 2.21 (95% CI:1.32-3.82) and 2.18 (95% CI:1.69-2.81), respectively. Moreover, compared to the participants in the NTNW group, the ORs of the decreased ankle-brachial index (ABI) and elevated Brachial-ankle pulse wave velocity (baPWV) were 2.24 (95% CI:1.38-3.80) and 1.63 (95% CI:1.28-2.06) in the HTGW group. Conclusion In summary, there was an association between hyper-triglyceridemic waist phenotype and diabetic vascular complications in the Chinese population.
Collapse
Affiliation(s)
- Kun Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Bin Cao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| |
Collapse
|
12
|
Zou Y, Zhao L, Zhang J, Wang Y, Wu Y, Ren H, Wang T, Zhao Y, Xu H, Li L, Tong N, Liu F. Metabolic-associated fatty liver disease increases the risk of end-stage renal disease in patients with biopsy-confirmed diabetic nephropathy: a propensity-matched cohort study. Acta Diabetol 2023; 60:225-233. [PMID: 36319797 DOI: 10.1007/s00592-022-01978-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/18/2022] [Indexed: 01/21/2023]
Abstract
AIMS To investigate the relationship between metabolic-associated fatty liver disease (MAFLD) and end-stage renal disease (ESRD) in patients with biopsy-confirmed diabetic nephropathy (DN). METHODS A total of 316 participants with biopsy-confirmed DN between January 2008 and December 2019 were retrospectively assessed. Kaplan-Meier curve and Cox proportional hazard models were used to compare the risk of incident ESRD in 50 patients with MAFLD and 50 patients without MAFLD, after using propensity score matching (PSM) to address the imbalances of sex, age, baseline-estimated glomerular filtration rate, serum albumin, 24-h urine protein, hemoglobin and systolic blood pressure. RESULTS During the median follow-up period of 3 years, there were 19 ESRD outcome events (19%) in PSM cohort. Kaplan-Meier curve analysis suggested that renal survival significantly deteriorated in patients with MAFLD versus those without MAFLD (p = 0.021). Additionally, the hazard ratios (95% confidence interval) of MAFLD were 3.12 (1.09-8.95, p = 0.035), 3.36 (1.09-10.43, p = 0.036), 3.66 (1.22-10.98, p = 0.021), 4.25 (1.34-13.45, p = 0.014), 3.11 (1.08-8.96, p = 0.035) and 5.84 (1.94-18.5, p = 0.003) after adjustment for six models, including demographic, clinical and pathological characteristics as well as medication use at the time of renal biopsy, respectively. Besides, patients with higher liver fibrosis score had a greater possibility of ESRD, comparing to those with lower liver fibrosis score (p = 0.002). CONCLUSIONS MAFLD increases the risk of incident ESRD in patients with biopsy-proven DN. Further research is needed to determine whether treatment targeting MAFLD improves the prognosis of DN.
Collapse
Affiliation(s)
- Yutong Zou
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Lijun Zhao
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Junlin Zhang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yiting Wang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yucheng Wu
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Honghong Ren
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Tingli Wang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yuancheng Zhao
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Huan Xu
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lin Li
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Nanwei Tong
- Division of Endocrinology, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
- Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Fang Liu
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
13
|
Swarbrick MM, Cox CL, Graham JL, Knudsen LB, Stanhope K, Raun K, Havel PJ. Growth hormone treatment does not augment the anti-diabetic effects of liraglutide in UCD-T2DM rats. Endocrinol Diabetes Metab 2022; 6:e392. [PMID: 36480511 PMCID: PMC9836246 DOI: 10.1002/edm2.392] [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: 08/09/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The incretin hormone glucagon-like peptide-1 (GLP-1) slows gastric emptying, increases satiety and enhances insulin secretion. GLP-1 receptor agonists, such as liraglutide, are used therapeutically in humans to improve glycaemic control and delay the onset of type 2 diabetes mellitus (T2DM). In UCD-T2DM rats, a model of polygenic obesity and insulin resistance, we have previously reported that daily liraglutide administration delayed diabetes onset by >4 months. Growth hormone (GH) may exert anti-diabetic effects, including increasing β-cell mass and insulin secretion, while disrupting GH signalling in mice reduces both the size and number of pancreatic islets. We therefore hypothesized that GH supplementation would augment liraglutide's anti-diabetic effects. METHODS Male UCD-T2DM rats were treated daily with GH (0.3 mg/kg) and/or liraglutide (0.2 mg/kg) from 2 months of age. Control (vehicle) and food-restricted (with food intake matched to liraglutide-treated rats) groups were also studied. The effects of treatment on diabetes onset and weight gain were assessed, as well as measures of glucose tolerance, lipids and islet morphology. RESULTS Liraglutide treatment significantly reduced food intake and body weight and improved glucose tolerance and insulin sensitivity, relative to controls. After 4.5 months, none of the liraglutide-treated rats had developed T2DM (overall p = .019). Liraglutide-treated rats also displayed lower fasting triglyceride (TG) concentrations and lower hepatic TG content, compared to control rats. Islet morphology was improved in liraglutide-treated rats, with significantly increased pancreatic insulin content (p < .05 vs. controls). Although GH treatment tended to increase body weight (and gastrocnemius muscle weight), there were no obvious effects on diabetes onset or other diabetes-related outcomes. CONCLUSION GH supplementation did not augment the anti-diabetic effects of liraglutide.
Collapse
Affiliation(s)
- Michael M. Swarbrick
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA,Present address:
Bone Research Program, ANZAC Research InstituteThe University of SydneyConcordNew South WalesAustralia,Present address:
Concord Clinical School, Faculty of Medicine and HealthThe University of SydneyAustralia
| | - Chad L. Cox
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| | - James L. Graham
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| | | | - Kimber Stanhope
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| | | | - Peter J. Havel
- Departments of Nutrition and Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisOne Shielad AvenueDavisCaliforniaUSA
| |
Collapse
|
14
|
Wang Y, Liu Y, Yang R, Li Z, Su J, Yang T, Ma M, Pan G, Wang X, Li L, Yu C. Remnant cholesterol for the detection of glucose metabolic states in patients with coronary heart disease angina pectoris. Acta Diabetol 2022; 59:1339-1347. [PMID: 35871108 DOI: 10.1007/s00592-022-01935-7] [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: 05/22/2022] [Accepted: 06/29/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND This study aimed to evaluate the relationship between remnant cholesterol (RC) and glucose metabolic states in coronary heart disease (CHD) patients with angina pectoris. METHODS This study collected data from 11,557 CHD patients with angina pectoris aged 35-75 years in Tianjin, China. Participants were divided into normal glucose regulation (NGR), prediabetes (Pre-DM) and diabetes mellitus (DM) groups according to glucose metabolic states. Linear regression analysis was used to explore the relationship between glucose metabolism [fasting blood glucose (FBG) and glycated hemoglobin (HbA1c)] and RC levels. Logistic regression was performed to analyze the relationship between RC levels and glucose metabolic states. RESULTS Among all participants, 5883 (50.9%) had a DM state and 4034 (34.9%) had a Pre-DM state. FBG levels and HbA1c levels were positively related with RC in all patients (P < 0.001). NGR was used as a reference, multi-adjusted model showing that RC level was significantly associated with Pre-DM [Odds ratio (OR): 1.37; 95% confidence interval (CI) 1.19-1.56; P < 0.001] and DM state (OR:1.47; 95% CI 1.29-1.67; P < 0.001). When considering RC as categorical variables (tertiles), using T1 as a reference, T3 had the strongest relationship between RC levels and Pre-DM and DM state in univariate model and multivariate model. In the stratified analyses, the association between RC levels and pre-DM and DM in women was higher than that in men, and the elderly patients was higher than in the middle-aged patients. CONCLUSION The study demonstrated a significant association between RC levels and pre-DM and DM state among CHD patients with angina pectoris, and the relationship was stronger in women and elderly patients.
Collapse
Affiliation(s)
- Yang Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yijia Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Rongrong Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jinyu Su
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tong Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Mei Ma
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Guangwei Pan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China.
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
15
|
Caffeic Acid Phenethyl Ester Inhibits Basal Lipolysis by Activating PPAR-Gamma and Increasing Lipid Droplet-Associated Perilipin in Mature Rat Adipocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6007233. [PMID: 36082179 PMCID: PMC9448551 DOI: 10.1155/2022/6007233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
Abstract
Abnormal lipolysis is correlated with metabolic syndrome. Caffeic acid phenethyl ester (CAPE), a natural product from honeybee hives, has been reported to improve metabolic syndrome. However, the effects of CAPE on lipolysis and perilipin-1 (the major lipid droplet-associated protein) in mature adipocytes were not clarified. In this study, mature adipocytes were isolated from the epididymal fat pads of male rats and incubated with CAPE to estimate lipolysis by measuring glycerol release. It was found that the basal lipolysis was inhibited by CAPE in a dose- and time-dependent manner. The lipid droplet-associated perilipin-1 and phosphorylated peroxisome proliferator-activated receptor (PPAR) gamma levels increased following CAPE treatment by Western blot analysis. Moreover, a specific PPAR-gamma inhibitor (T0070907) could partly reverse the effect of CAPE on basal lipolysis. Furthermore, treatment of adipocytes with dibutyryl-cAMP (db-cAMP) or isoproterenol (ISO) increased lipolysis, but the drug-induced lipolysis was abrogated by combination treatment with CAPE. The lipid droplet-associated perilipin-1 level was also decreased in the drug-induced groups but increased when combined treatment with CAPE. In conclusion, our results revealed that a decrease in basal lipolysis and an increase in lipid droplet-associated perilipin-1 levels induced by CAPE may be involved in the regulation of lipid metabolism through activation of PPAR-gamma in mature adipocytes.
Collapse
|
16
|
Lee IS, Ko SJ, Lee YN, Lee G, Rahman MH, Kim B. The Effect of Laminaria japonica on Metabolic Syndrome: A Systematic Review of Its Efficacy and Mechanism of Action. Nutrients 2022; 14:3046. [PMID: 35893900 PMCID: PMC9370431 DOI: 10.3390/nu14153046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/10/2022] Open
Abstract
Metabolic syndrome (MetS) is a medical condition characterized by abdominal obesity, insulin resistance, high blood pressure, and hyperlipidemia. An increase in the incidence of MetS provokes an escalation in health care costs and a downturn in quality of life. However, there is currently no cure for MetS, and the absence of immediate treatment for MetS has prompted the development of novel therapies. In accordance with recent studies, the brown seaweed Laminaria japonica (LJP) has anti-inflammatory and antioxidant properties, and so forth. LJP contains bioactive compounds used as food globally, and it has been used as a medicine in East Asian countries. We conducted a systematic review to examine whether LJP could potentially be a useful therapeutic drug for MetS. The following databases were searched from initiation to September 2021: PubMed, Web of Science, EMBASE, and Cochrane Central Register of Controlled Trials Library. Clinical trials and in vivo studies evaluating the effects of LJP on MetS were included. LJP reduces the oxidative stress-related lipid mechanisms, inflammatory cytokines and macrophage-related chemokines, muscle cell proliferation, and migration. Bioactive-glucosidase inhibitors reduce diabetic complications, a therapeutic target in obesity and type 2 diabetes. In obesity, LJP increases AMP-activated protein kinase and decreases acetyl-CoA carboxylase. Based on our findings, we suggest that LJP could treat MetS, as it has pharmacological effects on MetS.
Collapse
Affiliation(s)
- In-Seon Lee
- Department of Meridians and Acupoints, College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea;
- Acupuncture & Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea
| | - Seok-Jae Ko
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea;
| | - Yu Na Lee
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| | - Gahyun Lee
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| | - Md. Hasanur Rahman
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| |
Collapse
|
17
|
Biondi G, Marrano N, Borrelli A, Rella M, Palma G, Calderoni I, Siciliano E, Lops P, Giorgino F, Natalicchio A. Adipose Tissue Secretion Pattern Influences β-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23105522. [PMID: 35628332 PMCID: PMC9143684 DOI: 10.3390/ijms23105522] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
The dysregulation of the β-cell functional mass, which is a reduction in the number of β-cells and their ability to secure adequate insulin secretion, represents a key mechanistic factor leading to the onset of type 2 diabetes (T2D). Obesity is recognised as a leading cause of β-cell loss and dysfunction and a risk factor for T2D. The natural history of β-cell failure in obesity-induced T2D can be divided into three steps: (1) β-cell compensatory hyperplasia and insulin hypersecretion, (2) insulin secretory dysfunction, and (3) loss of β-cell mass. Adipose tissue (AT) secretes many hormones/cytokines (adipokines) and fatty acids that can directly influence β-cell function and viability. As this secretory pattern is altered in obese and diabetic patients, it is expected that the cross-talk between AT and pancreatic β-cells could drive the maintenance of the β-cell integrity under physiological conditions and contribute to the reduction in the β-cell functional mass in a dysmetabolic state. In the current review, we summarise the evidence of the ability of the AT secretome to influence each step of β-cell failure, and attempt to draw a timeline of the alterations in the adipokine secretion pattern in the transition from obesity to T2D that reflects the progressive deterioration of the β-cell functional mass.
Collapse
|
18
|
Zheng X, Ho QWC, Chua M, Stelmashenko O, Yeo XY, Muralidharan S, Torta F, Chew EGY, Lian MM, Foo JN, Jung S, Wong SH, Tan NS, Tong N, Rutter GA, Wenk MR, Silver DL, Berggren PO, Ali Y. Destabilization of β Cell FIT2 by saturated fatty acids alter lipid droplet numbers and contribute to ER stress and diabetes. Proc Natl Acad Sci U S A 2022; 119:e2113074119. [PMID: 35254894 PMCID: PMC8931238 DOI: 10.1073/pnas.2113074119] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/29/2022] [Indexed: 02/05/2023] Open
Abstract
SignificanceWith obesity on the rise, there is a growing appreciation for intracellular lipid droplet (LD) regulation. Here, we show how saturated fatty acids (SFAs) reduce fat storage-inducing transmembrane protein 2 (FIT2)-facilitated, pancreatic β cell LD biogenesis, which in turn induces β cell dysfunction and death, leading to diabetes. This mechanism involves direct acylation of FIT2 cysteine residues, which then marks the FIT2 protein for endoplasmic reticulum (ER)-associated degradation. Loss of β cell FIT2 and LDs reduces insulin secretion, increases intracellular ceramides, stimulates ER stress, and exacerbates diet-induced diabetes in mice. While palmitate and stearate degrade FIT2, unsaturated fatty acids such as palmitoleate and oleate do not, results of which extend to nutrition and diabetes.
Collapse
Affiliation(s)
- Xiaofeng Zheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Qing Wei Calvin Ho
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
| | - Minni Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
| | - Olga Stelmashenko
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
| | - Xin Yi Yeo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138667, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, S119228, Singapore
| | - Sneha Muralidharan
- Singapore Lipidomics Incubator, Department of Medicine, National University of Singapore, S117456, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Department of Biochemistry, Life Sciences Institute and Yong Loo Lin School of Medicine, National University of Singapore, S117456, Singapore
| | - Elaine Guo Yan Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Human Genetics, A*STAR, Genome Institute of Singapore, S138672, Singapore
| | - Michelle Mulan Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Human Genetics, A*STAR, Genome Institute of Singapore, S138672, Singapore
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Human Genetics, A*STAR, Genome Institute of Singapore, S138672, Singapore
| | - Sangyong Jung
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138667, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, S117593, Singapore
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, S637551, Singapore
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Guy A. Rutter
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London SW7 2AZ, United Kingdom
- Le Centre de recherche du Centre hospitalier de l’Université de Montréal (CR-CHUM), University of Montréal, Montréal, QC H2X 0A9, Canada
| | - Markus R. Wenk
- Singapore Lipidomics Incubator, Department of Biochemistry, Life Sciences Institute and Yong Loo Lin School of Medicine, National University of Singapore, S117456, Singapore
| | - David L. Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke–National University of Singapore Graduate Medical School, S169857, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
| |
Collapse
|
19
|
Metabolic Syndrome: Updates on Pathophysiology and Management in 2021. Int J Mol Sci 2022; 23:ijms23020786. [PMID: 35054972 PMCID: PMC8775991 DOI: 10.3390/ijms23020786] [Citation(s) in RCA: 587] [Impact Index Per Article: 195.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolic syndrome (MetS) forms a cluster of metabolic dysregulations including insulin resistance, atherogenic dyslipidemia, central obesity, and hypertension. The pathogenesis of MetS encompasses multiple genetic and acquired entities that fall under the umbrella of insulin resistance and chronic low-grade inflammation. If left untreated, MetS is significantly associated with an increased risk of developing diabetes and cardiovascular diseases (CVDs). Given that CVDs constitute by far the leading cause of morbidity and mortality worldwide, it has become essential to investigate the role played by MetS in this context to reduce the heavy burden of the disease. As such, and while MetS relatively constitutes a novel clinical entity, the extent of research about the disease has been exponentially growing in the past few decades. However, many aspects of this clinical entity are still not completely understood, and many questions remain unanswered to date. In this review, we provide a historical background and highlight the epidemiology of MetS. We also discuss the current and latest knowledge about the histopathology and pathophysiology of the disease. Finally, we summarize the most recent updates about the management and the prevention of this clinical syndrome.
Collapse
|
20
|
Iacob SA, Iacob DG. Non-Alcoholic Fatty Liver Disease in HIV/HBV Patients - a Metabolic Imbalance Aggravated by Antiretroviral Therapy and Perpetuated by the Hepatokine/Adipokine Axis Breakdown. Front Endocrinol (Lausanne) 2022; 13:814209. [PMID: 35355551 PMCID: PMC8959898 DOI: 10.3389/fendo.2022.814209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is strongly associated with the metabolic syndrome and is one of the most prevalent comorbidities in HIV and HBV infected patients. HIV plays an early and direct role in the development of metabolic syndrome by disrupting the mechanism of adipogenesis and synthesis of adipokines. Adipokines, molecules that regulate the lipid metabolism, also contribute to the progression of NAFLD either directly or via hepatic organokines (hepatokines). Most hepatokines play a direct role in lipid homeostasis and liver inflammation but their role in the evolution of NAFLD is not well defined. The role of HBV in the pathogenesis of NAFLD is controversial. HBV has been previously associated with a decreased level of triglycerides and with a protective role against the development of steatosis and metabolic syndrome. At the same time HBV displays a high fibrogenetic and oncogenetic potential. In the HIV/HBV co-infection, the metabolic changes are initiated by mitochondrial dysfunction as well as by the fatty overload of the liver, two interconnected mechanisms. The evolution of NAFLD is further perpetuated by the inflammatory response to these viral agents and by the variable toxicity of the antiretroviral therapy. The current article discusses the pathogenic changes and the contribution of the hepatokine/adipokine axis in the development of NAFLD as well as the implications of HIV and HBV infection in the breakdown of the hepatokine/adipokine axis and NAFLD progression.
Collapse
Affiliation(s)
- Simona Alexandra Iacob
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Infectious Diseases, National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| | - Diana Gabriela Iacob
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Infectious Diseases, Emergency University Hospital, Bucharest, Romania
- *Correspondence: Diana Gabriela Iacob,
| |
Collapse
|
21
|
de Melo FTC, Felício KM, de Queiroz NNM, de Rider Brito HA, Neto JFA, Janaú LC, de Souza Neto NJK, Silva ALA, de Lemos MN, de Oliveira MCNI, de Alcântara AL, de Moraes LV, de Souza ÍJA, Said NM, da Silva WM, de Lemos GN, Dos Santos MC, De Souza D Albuquerque Silva L, Motta ARB, de Figueiredo PBB, de Souza ACCB, Piani PPF, Felício JS. High-dose Vitamin D Supplementation on Type 1 Diabetes Mellitus Patients: Is there an Improvement in Glycemic Control? Curr Diabetes Rev 2022; 18:e010521189964. [PMID: 33413064 DOI: 10.2174/1573399817666210106102643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Some authors evaluated the effect of VD on hyperglycemia in T1DM, but the results remain controversial. This study aims to analyze the effects of high-dose VD supplementation on T1DM patients' glycemic levels, maintaining stable doses of insulin. METHODS Prospective, 12-week clinical trial including 67 T1DM patients, supplemented with high doses of cholecalciferol according to participants' VD value. Patients with VD levels below 30 ng/mL received 10,000 IU/day; those with levels between 30-60 ng/mL received 4,000 IU/day. Patients who had not achieved 25(OH)D levels > 30 ng/ml or presented insulin dose variation during the study were not analyzed. RESULTS Only 46 out of 67 patients accomplished the criteria at the end of the study. There was no general improvement in the glycemic control evaluated by HbA1c (9.4 ± 2.4 vs 9.4 ± 2.6, p=NS) after VD supplementation. However, a post-hoc analysis, based on HbA1c variation, identified patients who had HbA1c reduced at least 0.6% (group 1, N = 13 (28%)). In addition, a correlation between 25(OH)D levels with HbA1c and total insulin dose at the end of the study was observed (r = -0.3, p<0.05; r=-0.4, p<0.05, respectively), and a regression model demonstrated that 25(OH)D was independent of BMI, duration of T1DM and final total insulin dose, being capable of determining 9.2% of HbA1c final levels (Unstandardized B coefficient = -0.033 (CI 95%: -0.064 to -0.002), r2 = 0.1, p <0.05). CONCLUSION Our data suggest that VD is not widely recommended for glycemic control. Nevertheless, specific patients might benefit from this approach.
Collapse
Affiliation(s)
- Franciane Trindade Cunha de Melo
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Karem Mileo Felício
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Natércia Neves Marques de Queiroz
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Hana Andrade de Rider Brito
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - João Felício Abrahão Neto
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Luísa Corrêa Janaú
- State University of Pará, Platter Perebebuí, 2623, Marco, Belém, Pará,Brazil
| | - Norberto Jorge Kzan de Souza Neto
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Ana Luíza Aires Silva
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Manuela Nascimento de Lemos
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Maria Clara Neres Iunes de Oliveira
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Angélica Leite de Alcântara
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Lorena Vilhena de Moraes
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Ícaro José Araújo de Souza
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Nivin Mazen Said
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Wanderson Maia da Silva
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Gabriela Nascimento de Lemos
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Márcia Costa Dos Santos
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Lilian De Souza D Albuquerque Silva
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Ana Regina Bastos Motta
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | | | - Ana Carolina Contente Braga de Souza
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - Pedro Paulo Freire Piani
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| | - João Soares Felício
- University Hospital João de Barros Barreto, Federal University of Pará, Endocrinology Division, Mundurucus Street, 4487, Guamá, Belém, Pará,Brazil
| |
Collapse
|
22
|
Lim SH, Lee HS, Han HK, Choi CI. Saikosaponin A and D Inhibit Adipogenesis via the AMPK and MAPK Signaling Pathways in 3T3-L1 Adipocytes. Int J Mol Sci 2021; 22:ijms222111409. [PMID: 34768840 PMCID: PMC8583978 DOI: 10.3390/ijms222111409] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/07/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a lipid metabolism disorder caused by genetic, medicinal, nutritional, and other environmental factors. It is characterized by a complex condition of excess lipid accumulation in adipocytes. Adipogenesis is a differentiation process that converts preadipocytes into mature adipocytes and contributes to excessive fat deposition. Saikosaponin A (SSA) and saikosaponin D (SSD) are triterpenoid saponins separated from the root of the Bupleurum chinensis, which has long been used to treat inflammation, fever, and liver diseases. However, the effects of these constituents on lipid accumulation and obesity are poorly understood. We investigated the anti-obesity effects of SSA and SSD in mouse 3T3-L1 adipocytes. The MTT assay was performed to measure cell viability, and Oil Red O staining was conducted to determine lipid accumulation. Various adipogenic transcription factors were evaluated at the protein and mRNA levels by Western blot assay and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Here, we showed that SSA and SSD significantly inhibited lipid accumulation without affecting cell viability within the range of the tested concentrations (0.938–15 µM). SSA and SSD also dose-dependently suppressed the expression of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein alpha (C/EBPα), sterol regulatory element binding protein-1c (SREBP-1c), and adiponectin. Furthermore, the decrease of these transcriptional factors resulted in the repressed expression of several lipogenic genes including fatty acid binding protein (FABP4), fatty acid synthase (FAS), and lipoprotein lipase (LPL). In addition, SSA and SSD enhanced the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and its substrate, acetyl-CoA carboxylase (ACC), and inhibited the phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) and p38, but not c-Jun-N-terminal kinase (JNK). These results suggest that SSA and SSD inhibit adipogenesis through the AMPK or mitogen-activated protein kinase (MAPK) pathways in the early stages of adipocyte differentiation. This is the first study on the anti-adipogenic effects of SSA and SSD, and further research in animals and humans is necessary to confirm the potential of saikosaponins as therapeutic agents for obesity.
Collapse
Affiliation(s)
- Sung Ho Lim
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea; (S.H.L.); (H.S.L.)
| | - Ho Seon Lee
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea; (S.H.L.); (H.S.L.)
| | - Hyo-Kyung Han
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea;
| | - Chang-Ik Choi
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea; (S.H.L.); (H.S.L.)
- Correspondence: ; Tel.: +82-31-961-5230
| |
Collapse
|
23
|
Govatati S, Pichavaram P, Mani AM, Kumar R, Sharma D, Dienel A, Meena S, Puchowicz MA, Park EA, Rao GN. Novel role of xanthine oxidase-dependent H 2O 2 production in 12/15-lipoxygenase-mediated de novo lipogenesis, triglyceride biosynthesis and weight gain. Redox Biol 2021; 47:102163. [PMID: 34655995 PMCID: PMC8577505 DOI: 10.1016/j.redox.2021.102163] [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: 09/16/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 11/16/2022] Open
Abstract
12/15-lipoxygenase (12/15-LOX) plays an essential role in oxidative conversion of polyunsaturated fatty acids into various bioactive lipid molecules. Although 12/15-LOX's role in the pathophysiology of various human diseases has been well studied, its role in weight gain is controversial and poorly clarified. Here, we demonstrated the role of 12/15-LOX in high-fat diet (HFD)-induced weight gain in a mouse model. We found that 12/15-LOX mediates HFD-induced de novo lipogenesis (DNL), triglyceride (TG) biosynthesis and the transport of TGs from the liver to adipose tissue leading to white adipose tissue (WAT) expansion and weight gain via xanthine oxidase (XO)-dependent production of H2O2. 12/15-LOX deficiency leads to cullin2-mediated ubiquitination and degradation of XO, thereby suppressing H2O2 production, DNL and TG biosynthesis resulting in reduced WAT expansion and weight gain. These findings infer that manipulation of 12/15-LOX metabolism may manifest a potential therapeutic target for weight gain and obesity. 12/15-LOX-12(S)-HETE axis via activation of CREB-Egr1 enhances TG biosynthesis. 12/15-LOX-12(S)-HETE axis via activation of SREBP1c triggers DNL. H2O2 mediates 12/15-LOX-12(S)-HETE axis-induced DNL and TG biosynthesis. 12/15-LOX via TG biosynthesis leads to WAT expansion and body weight gain. Downstream to 12/15-LOX, H2O2-mediates WAT expansion and body weight gain.
Collapse
Affiliation(s)
- Suresh Govatati
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Prahalathan Pichavaram
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Arul M Mani
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Deepti Sharma
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Ari Dienel
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Sunita Meena
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Edwards A Park
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
24
|
Zhou B, Zhang Y, Li S, Wu L, Fejes-Toth G, Naray-Fejes-Toth A, Soukas AA. Serum- and glucocorticoid-induced kinase drives hepatic insulin resistance by directly inhibiting AMP-activated protein kinase. Cell Rep 2021; 37:109785. [PMID: 34610303 PMCID: PMC8576737 DOI: 10.1016/j.celrep.2021.109785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/05/2021] [Accepted: 09/10/2021] [Indexed: 12/01/2022] Open
Abstract
A hallmark of type 2 diabetes (T2D) is hepatic resistance to insulin's glucose-lowering effects. The serum- and glucocorticoid-regulated family of protein kinases (SGK) is activated downstream of mechanistic target of rapamycin complex 2 (mTORC2) in response to insulin in parallel to AKT. Surprisingly, despite an identical substrate recognition motif to AKT, which drives insulin sensitivity, pathological accumulation of SGK1 drives insulin resistance. Liver-specific Sgk1-knockout (Sgk1Lko) mice display improved glucose tolerance and insulin sensitivity and are protected from hepatic steatosis when fed a high-fat diet. Sgk1 promotes insulin resistance by inactivating AMP-activated protein kinase (AMPK) via phosphorylation on inhibitory site AMPKαSer485/491. We demonstrate that SGK1 is dominant among SGK family kinases in regulation of insulin sensitivity, as Sgk1, Sgk2, and Sgk3 triple-knockout mice have similar increases in hepatic insulin sensitivity. In aggregate, these data suggest that targeting hepatic SGK1 may have therapeutic potential in T2D.
Collapse
Affiliation(s)
- Ben Zhou
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yuyao Zhang
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Sainan Li
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Lianfeng Wu
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Geza Fejes-Toth
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Aniko Naray-Fejes-Toth
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Alexander A Soukas
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
25
|
Malinska H, Klementová M, Kudlackova M, Veleba J, Hoskova E, Oliyarnyk O, Markova I, Thieme L, Hill M, Pelikanova T, Kahleova H. A plant-based meal reduces postprandial oxidative and dicarbonyl stress in men with diabetes or obesity compared with an energy- and macronutrient-matched conventional meal in a randomized crossover study. Nutr Metab (Lond) 2021; 18:84. [PMID: 34507586 PMCID: PMC8434736 DOI: 10.1186/s12986-021-00609-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/28/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Increased oxidative/dicarbonyl stress and chronic inflammation are considered key pathophysiological mediators in the progression of complications in obesity and type 2 diabetes (T2D). Lifestyle and diet composition have a major impact. In this study, we tested the effects of a vegan (V) and a conventional meat containg (M) meal, matched for energy and macronutrients, on postprandial oxidative and dicarbonyl stress, inflammatory markers and appetite hormones. METHODS A randomised crossover design was used to evaluate T2D, obese with normal glucose tolerance and control participants (n = 20 in each group), with serum concentrations of analytes determined at 0, 120 and 180 min. Repeated-measures ANOVA was used for statistical analysis. RESULTS In T2D subjects, we observed decreased postprandial concentrations of oxidised glutathione (p ˂ 0.001) and increased glutathione peroxidase activity (p = 0.045) after the V-meal consumption, compared with the M-meal. In obese participants, V-meal consumption increased postprandial concentrations of reduced glutathione (p = 0.041) and decreased methylglyoxal concentrations (p = 0.023). There were no differences in postprandial secretion of TNFα, MCP-1 or ghrelin in T2D or obese men, but we did observe higher postprandial secretion of leptin after the V-meal in T2D men (p = 0.002) compared with the M-meal. CONCLUSIONS The results show that a plant-based meal is efficient in ameliorating the postprandial oxidative and dicarbonyl stress compared to a conventional energy- and macronutrient-matched meal, indicating the therapeutic potential of plant-based nutrition in improving the progression of complications in T2D and obese patients. Registered under ClinicalTrials.gov Identifier No. NCT02474147.
Collapse
Affiliation(s)
- Hana Malinska
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marta Klementová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Jiri Veleba
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Hoskova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Olena Oliyarnyk
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Irena Markova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lenka Thieme
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Hill
- Institute of Endocrinology, Prague, Czech Republic
| | - Terezie Pelikanova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Hana Kahleova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic. .,Physicians Committee for Responsible Medicine, 5100 Wisconsin Ave, NW, Suite 400, Washington, DC, 20016, USA.
| |
Collapse
|
26
|
Yang J, Zhou S, Gu Z, Cheng L, Cui C, Shen Y, Hong Y. Effect of starch-hydrocolloid complexes with heat-moisture treatment on in vivo digestibility. Food Funct 2021; 12:8017-8025. [PMID: 34269784 DOI: 10.1039/d1fo01586a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The purpose of this study was to investigate the effect of starch-hydrocolloid (gum arabic, xanthan gum, and guar gum) complexes with heat-moisture treatment (HMT) on in vivo digestibility. In vivo digestibility experiments revealed that the body weight, liver weight, and fat index of mice in the intervention group were significantly reduced compared with those in the high-fat group. Glucose tolerance improved, and blood lipid levels, liver and adipose tissue morphology returned to normal. The results of mRNA expression levels showed that the intervention of corn starch-hydrocolloid complexes after HMT down-regulated the expression level of genes related to fat synthesis compared with the high-fat group, which could decrease lipid deposition and stabilize blood lipid levels. Results revealed that starch-xanthan gum complex (1 : 40 ratio) with HMT could markedly reduce the digestibility of starch. Overall, this study provides new ideas for the application of low-glycemic-index and functional foods.
Collapse
Affiliation(s)
- Jie Yang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Qie R, Li Q, Zhao Y, Han M, Liu D, Guo C, Zhou Q, Tian G, Huang S, Wu X, Zhang Y, Qin P, Li H, Wang J, Cheng R, Lin J, Sun X, Wu Y, Li Y, Yang X, Zhao Y, Feng Y, Zhang M, Hu D. Association of hypertriglyceridemic waist-to-height ratio and its dynamic status with risk of type 2 diabetes mellitus: The Rural Chinese Cohort Study. Diabetes Res Clin Pract 2021; 179:108997. [PMID: 34371063 DOI: 10.1016/j.diabres.2021.108997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022]
Abstract
AIMS To evaluate the risk of type 2 diabetes mellitus (T2DM) in a prospective study with hypertriglyceridemic waist-to-height ratio (HWHtR) and its dynamic status. METHODS We collected data for 12,248 participants ≥18 years in this study. Cox's proportional-hazards regression was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for T2DM risk by baseline HWHtR. Multiple logistic regression analysis was used to estimate odds ratios (ORs) and 95% CIs for T2DM risk by transformation in HWHtR. RESULTS We identified 839 T2DM cases during a median follow-up of 5.92 years. Compared with normal TG level and normal WHtR, T2DM risk was increased with high TG level and high WHtR (aHR 2.04, 95% CI 1.49-2.79). Similar results were observed in subgroup analyses by sex and age. During follow-up, T2DM risk was increased with stable high TG level and high WHtR (aOR 4.45, 95% CI 2.76-7.17) compared with stable normal TG level and normal WHtR. The results above were robust in sensitivity analyses. CONCLUSIONS HWHtR phenotype and its dynamic status were associated with risk of T2DM. Our study suggests that primary prevention and avoiding the appearance of the HWHtR phenotype in the rural Chinese population may reduce the T2DM risk.
Collapse
Affiliation(s)
- Ranran Qie
- Department of Endocrinology, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, PR China.; Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Quanman Li
- Department of Endocrinology, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, PR China.; Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yang Zhao
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Minghui Han
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Dechen Liu
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Chunmei Guo
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Qionggui Zhou
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Gang Tian
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shengbing Huang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xiaoyan Wu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yanyan Zhang
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Pei Qin
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Honghui Li
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Jian Wang
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ruirong Cheng
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Jinchun Lin
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xizhuo Sun
- Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yuying Wu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yang Li
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xingjin Yang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yifei Feng
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Ming Zhang
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Dongsheng Hu
- Department of Endocrinology, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, PR China.; Study Team of Shenzhen's Sanming Project, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
28
|
PEGylated AdipoRon derivatives improve glucose and lipid metabolism under insulinopenic and high-fat diet conditions. J Lipid Res 2021; 62:100095. [PMID: 34214600 PMCID: PMC8327158 DOI: 10.1016/j.jlr.2021.100095] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
The pleiotropic actions of adiponectin in improving cell survival and metabolism have motivated the development of small-molecule therapeutic agents for treating diabetes and lipotoxicity. AdipoRon is a synthetic agonist of the adiponectin receptors, yet is limited by its poor solubility and bioavailability. In this work, we expand on the protective effects of AdipoRon in pancreatic β-cells and examine how structural modifications could affect the activity, pharmacokinetics, and bioavailability of this small molecule. We describe a series of AdipoRon analogs containing amphiphilic ethylene glycol (PEG) chains. Among these, AdipoRonPEG5 induced pleiotropic effects in mice under insulinopenic and high-fat diet (HFD) conditions. While both AdipoRon and AdipoRonPEG5 substantially attenuate palmitate-induced lipotoxicity in INS-1 cells, only AdipoRonPEG5 treatment is accompanied by a significant reduction in cytotoxic ceramides. In vivo, AdipoRonPEG5 can substantially reduce pancreatic, hepatic, and serum ceramide species, with a concomitant increase in the corresponding sphingoid bases and improves insulin sensitivity of mice under HFD feeding conditions. Furthermore, hyperglycemia in streptozotocin (STZ)-induced insulinopenic adiponectin-null mice is also attenuated upon AdipoRonPEG5 treatment. Our results suggest that AdipoRonPEG5 is more effective in reducing ceramides and dihydroceramides in the liver of HFD-fed mice than AdipoRon, consistent with its potent activity in activating ceramidase in vitro in INS-1 cells. Additionally, these results indicate that the beneficial effects of AdipoRonPEG5 can be partially attributed to improved pharmacokinetics as compared with AdipoRon, thus suggesting that further derivatization may improve affinity and tissue-specific targeting.
Collapse
|
29
|
Elumalai S, Karunakaran U, Moon JS, Won KC. NADPH Oxidase (NOX) Targeting in Diabetes: A Special Emphasis on Pancreatic β-Cell Dysfunction. Cells 2021; 10:cells10071573. [PMID: 34206537 PMCID: PMC8307876 DOI: 10.3390/cells10071573] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 12/24/2022] Open
Abstract
In type 2 diabetes, metabolic stress has a negative impact on pancreatic β-cell function and survival (T2D). Although the pathogenesis of metabolic stress is complex, an imbalance in redox homeostasis causes abnormal tissue damage and β-cell death due to low endogenous antioxidant expression levels in β-cells. Under diabetogenic conditions, the susceptibility of β-cells to oxidative damage by NADPH oxidase has been related to contributing to β-cell dysfunction. Here, we consider recent insights into how the redox response becomes deregulated under diabetic conditions by NADPH oxidase, as well as the therapeutic benefits of NOX inhibitors, which may provide clues for understanding the pathomechanisms and developing strategies aimed at the treatment or prevention of metabolic stress associated with β-cell failure.
Collapse
Affiliation(s)
- Suma Elumalai
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (S.E.); (U.K.)
| | - Udayakumar Karunakaran
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (S.E.); (U.K.)
| | - Jun-Sung Moon
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (S.E.); (U.K.)
- Department of Internal Medicine, Yeungnam Universtiy College of Medicine, Daegu 42415, Korea
- Correspondence: (J.-S.M.); (K.-C.W.); Tel.: +82-53-620-3825 (J.-S.W.); +82-53-620-3846 (K.-C.W.)
| | - Kyu-Chang Won
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (S.E.); (U.K.)
- Department of Internal Medicine, Yeungnam Universtiy College of Medicine, Daegu 42415, Korea
- Correspondence: (J.-S.M.); (K.-C.W.); Tel.: +82-53-620-3825 (J.-S.W.); +82-53-620-3846 (K.-C.W.)
| |
Collapse
|
30
|
Low expression of miR-19a-5p is associated with high mRNA expression of diacylglycerol O-acyltransferase 2 (DGAT2) in hybrid tilapia. Genomics 2021; 113:2392-2399. [PMID: 34022348 DOI: 10.1016/j.ygeno.2021.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/06/2021] [Accepted: 05/17/2021] [Indexed: 11/24/2022]
Abstract
DGAT2 (acyl CoA:diacylglycerol acyltransferase 2) is a key and rate-limiting enzyme that catalyzes the final step of triglyceride (TG) synthesis. In this study, hybrid tilapia were generated from Nile tilapia (♀) and blue tilapia (♂) crossing. The TG content levels in the liver of these tilapia were measured. The results showed that the TG content was higher in the hybrid tilapia. In addition, protein and mRNA expression levels in the tilapia livers were determined. Higher hepatic mRNA and protein expression of DGAT2 in the hybrid fish was found. A luciferase reporter assay with HEK293T cells revealed that miRNA-19a-5p targeted the 3'UTR of DGAT2, suggesting a direct regulatory mechanism. Using qRT-PCR, we found that DGAT2 mRNA levels had a negative correlation with miRNA-19a-5p expression in Nile tilapia and hybrid. Taken together, these findings provide evidence that miRNA-19a-5p is involved in TG synthesis in the regulation of lipid metabolism in tilapia.
Collapse
|
31
|
Becuwe M, Bond LM, Pinto AFM, Boland S, Mejhert N, Elliott SD, Cicconet M, Graham MM, Liu XN, Ilkayeva O, Saghatelian A, Walther TC, Farese RV. FIT2 is an acyl-coenzyme A diphosphatase crucial for endoplasmic reticulum homeostasis. J Cell Biol 2021; 219:152082. [PMID: 32915949 PMCID: PMC7659722 DOI: 10.1083/jcb.202006111] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum is a cellular hub of lipid metabolism, coordinating lipid synthesis with continuous changes in metabolic flux. Maintaining ER lipid homeostasis despite these fluctuations is crucial to cell function and viability. Here, we identify a novel mechanism that is crucial for normal ER lipid metabolism and protects the ER from dysfunction. We identify the molecular function of the evolutionarily conserved ER protein FIT2 as a fatty acyl–coenzyme A (CoA) diphosphatase that hydrolyzes fatty acyl–CoA to yield acyl 4′-phosphopantetheine. This activity of FIT2, which is predicted to be active in the ER lumen, is required in yeast and mammalian cells for maintaining ER structure, protecting against ER stress, and enabling normal lipid storage in lipid droplets. Our findings thus solve the long-standing mystery of the molecular function of FIT2 and highlight the maintenance of optimal fatty acyl–CoA levels as key to ER homeostasis.
Collapse
Affiliation(s)
- Michel Becuwe
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Laura M Bond
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Antonio F M Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA
| | - Sebastian Boland
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Niklas Mejhert
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Shane D Elliott
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA.,Howard Hughes Medical Institute, Boston, MA
| | - Marcelo Cicconet
- Image and Data Analysis Core, Harvard Medical School, Boston, MA
| | - Morven M Graham
- Center for Cellular and Molecular Imaging, Department of Cell Biology, Yale School of Medicine, New Haven, CT
| | - Xinran N Liu
- Center for Cellular and Molecular Imaging, Department of Cell Biology, Yale School of Medicine, New Haven, CT
| | - Olga Ilkayeva
- Departments of Medicine and Pharmacology and Cancer Biology, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA.,Howard Hughes Medical Institute, Boston, MA
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Cell Biology, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
32
|
Blair HR, Tomas C, Miwa S, Heath A, Russell A, Ginkel MV, Gunn D, Walker M. Peroxisomes and pancreatic beta-cell lipo-dysfunction. J Diabetes Complications 2021; 35:107843. [PMID: 33419633 DOI: 10.1016/j.jdiacomp.2020.107843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
AIMS Pancreatic beta-cell lipo-dysfunction decreases insulin secretion and predisposes to the development of type 2 diabetes. Through targeted Pex11β knockdown and peroxisome depletion, our aim was to investigate the specific contribution of peroxisomes to palmitate mediated pancreatic beta-cell dysfunction. METHODS MIN6 cells were transfected with probes targeted against Pex11β, a regulator of peroxisome abundance, or with scrambled control probes. Peroxisome abundance was measured by PMP-70 protein expression. 48 h post transfection, cells were incubated with 250 μM palmitate or BSA control for a further 48 h before measurement of glucose stimulated insulin secretion and of reactive oxygen species. RESULTS Pex11β knockdown decreased target gene expression by >80% compared with the scrambled control (P<0.001). This led to decreased PMP-70 expression (p<0.01) and a 22% decrease in peroxisome number (p<0.05). At 25 mM glucose, palmitate treatment decreased insulin secretion by 64% in the scrambled control cells (2.54±0.25 vs 7.07±0.83 [mean±SEM] ng/h/μg protein; Palmitate vs BSA P<0.001), but by just 37% in the Pex11β knockdown cells. Comparing responses in the presence of palmitate, insulin secretion at 25 mM glucose was significantly greater in the Pex11β knockdown cells compared with the scrambled controls (4.04±0.46 vs 2.54±0.25 ng/h/μg protein; p<0.05). Reactive oxygen species generation with palmitate was lower in the Pex11β knockdown cells compared with the scrambled controls (P<0.001). CONCLUSION Pex11β knockdown decreased peroxisome abundance, decreased palmitate mediated reactive oxygen species generation, and reversed the inhibitory effect of palmitate on insulin secretion. These findings reveal a distinct role of peroxisomes in palmitate mediated beta-cell dysfunction.
Collapse
Affiliation(s)
- Helen R Blair
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Cara Tomas
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Alan Heath
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Alison Russell
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | | | - David Gunn
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Mark Walker
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
33
|
Carvalho LSF, Benseñor IM, Nogueira ACC, Duncan BB, Schmidt MI, Blaha MJ, Toth PP, Jones SR, Santos RD, Lotufo PA, Sposito AC. Increased particle size of triacylglycerol-enriched remnant lipoproteins, but not their plasma concentration or lipid content, augments risk prediction of incident type 2 diabetes. Diabetologia 2021; 64:385-396. [PMID: 33159534 DOI: 10.1007/s00125-020-05322-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes prevention requires the accurate identification of those at high risk. Beyond the association of fasting serum triacylglycerols with diabetes, triacylglycerol-enriched remnant lipoproteins (TRLs) more accurately reflect pathophysiological changes that underlie progression to diabetes, such as hepatic insulin resistance, pancreatic steatosis and systemic inflammation. We hypothesised that TRL-related factors could improve risk prediction for incident diabetes. METHODS We included individuals from the Brazilian Longitudinal Study of Adult Health cohort. We trained a logistic regression model for the risk of incident diabetes in 80% of the cohort using tenfold cross-validation, and tested the model in the remaining 20% of the cohort (test set). Variables included medical history and traits of the metabolic syndrome, followed by TRL-related measurements (plasma concentration, TRL particle diameter, cholesterol and triacylglycerol content). TRL features were measured using NMR spectroscopy. Discrimination was assessed using the area under the receiver operating characteristic curve (AUROC) and the area under the precision-recall curve (AUPRC). RESULTS Among 4463 at-risk individuals, there were 366 new cases of diabetes after a mean (±SD) of 3.7 (±0.63) years of follow-up. We derived an 18-variable model with a global AUROC of 0.846 (95% CI: 0.829, 0.869). Overall TRL-related markers were not associated with diabetes. However, TRL particle diameter increased the AUROC, particularly in individuals with HbA1c <39 mmol/mol (5.7%) (hold-out test set [n = 659]; training-validation set [n = 2638]), but not in individuals with baseline HbA1c 39-46 mmol/mol (5.7-6.4%) (hold-out test set [n = 233]; training-validation set [n = 933]). In the subgroup with baseline HbA1c <39 mmol/mol (5.7%), AUROC in the test set increased from 0.717 (95% CI 0.603, 0.818) to 0.794 (95% CI 0.731, 0.862), and AUPRC in the test set rose from 0.582 to 0.701 when using the baseline model and the baseline model plus TRL particle diameter, respectively. TRL particle diameter was highly correlated with obesity, insulin resistance and inflammation in those with impaired fasting glucose at baseline, but less so in those with HbA1c <39 mmol/mol (5.7%). CONCLUSIONS/INTERPRETATION TRL particle diameter improves the prediction of diabetes, but only in individuals with HbA1c <39 mmol/mol (5.7%) at baseline. These data support TRL particle diameter as a risk factor that is changed early in the course of the pathophysiological processes that lead to the development of type 2 diabetes, even before glucose abnormalities are established. Graphical abstract.
Collapse
Affiliation(s)
- Luiz Sérgio F Carvalho
- Data Lab, Clarity Healthcare Intelligence, Jundiaí, SP, Brazil.
- Cardiology Division, Faculty of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil.
- Laboratory of Data for Quality of Care and Outcomes Research, Institute for Strategic Management in Healthcare DF (IGESDF), Brasília, DF, Brazil.
| | - Isabela M Benseñor
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo, SP, Brazil
| | - Ana C C Nogueira
- Laboratory of Data for Quality of Care and Outcomes Research, Institute for Strategic Management in Healthcare DF (IGESDF), Brasília, DF, Brazil
| | - Bruce B Duncan
- Postgraduate Studies Program in Epidemiology, School of Medicine and Hospital de Clínicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria I Schmidt
- Postgraduate Studies Program in Epidemiology, School of Medicine and Hospital de Clínicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Michael J Blaha
- The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Peter P Toth
- The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
- Preventive Cardiology, CGH Medical Center, Sterling, IL, USA
| | - Steven R Jones
- The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Raul D Santos
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo, SP, Brazil
- Lipid Clinic Heart Institute (InCor), University of São Paulo, Medical School Hospital, São Paulo, SP, Brazil
| | - Paulo A Lotufo
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo, SP, Brazil
| | - Andrei C Sposito
- Cardiology Division, Faculty of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil
| | | |
Collapse
|
34
|
Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. Biochem J 2020; 477:985-1008. [PMID: 32168372 DOI: 10.1042/bcj20190468] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/19/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
Fatty acids (FAs) are stored safely in the form of triacylglycerol (TAG) in lipid droplet (LD) organelles by professional storage cells called adipocytes. These lipids are mobilized during adipocyte lipolysis, the fundamental process of hydrolyzing TAG to FAs for internal or systemic energy use. Our understanding of adipocyte lipolysis has greatly increased over the past 50 years from a basic enzymatic process to a dynamic regulatory one, involving the assembly and disassembly of protein complexes on the surface of LDs. These dynamic interactions are regulated by hormonal signals such as catecholamines and insulin which have opposing effects on lipolysis. Upon stimulation, patatin-like phospholipase domain containing 2 (PNPLA2)/adipocyte triglyceride lipase (ATGL), the rate limiting enzyme for TAG hydrolysis, is activated by the interaction with its co-activator, alpha/beta hydrolase domain-containing protein 5 (ABHD5), which is normally bound to perilipin 1 (PLIN1). Recently identified negative regulators of lipolysis include G0/G1 switch gene 2 (G0S2) and PNPLA3 which interact with PNPLA2 and ABHD5, respectively. This review focuses on the dynamic protein-protein interactions involved in lipolysis and discusses some of the emerging concepts in the control of lipolysis that include allosteric regulation and protein turnover. Furthermore, recent research demonstrates that many of the proteins involved in adipocyte lipolysis are multifunctional enzymes and that lipolysis can mediate homeostatic metabolic signals at both the cellular and whole-body level to promote inter-organ communication. Finally, adipocyte lipolysis is involved in various diseases such as cancer, type 2 diabetes and fatty liver disease, and targeting adipocyte lipolysis is of therapeutic interest.
Collapse
|
35
|
Zhang Y, Qin P, Lou Y, Zhao P, Li X, Qie R, Wu X, Han M, Huang S, Zhao Y, Liu D, Wu Y, Li Y, Yang X, Zhao Y, Feng Y, Wang C, Ma J, Peng X, Chen H, Zhao D, Xu S, Wang L, Luo X, Zhang M, Hu D, Hu F. Association of TG/HDLC ratio trajectory and risk of type 2 diabetes: A retrospective cohort study in China. J Diabetes 2020; 13:402-412. [PMID: 33074586 DOI: 10.1111/1753-0407.13123] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/20/2020] [Accepted: 10/15/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The association of ratio of triglycerides to high-density lipoprotein cholesterol (TG/HDL-C ratio) change trajectory with risk of type 2 diabetes mellitus (T2DM) remains unknown. The aim of this study was to evaluate the association between risk of T2DM and TG/HDL-C ratio change trajectory. METHODS A total of 18 444 participants aged 18-80 years old were included in this cohort study. Linear regression and quadratic regression models were used to determine the TG/HDL-C ratio change trajectory. Logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between TG/HDL-C ratio change trajectory and probability of T2DM. RESULTS T2DM developed in 714 participants during a median follow-up of 5.74 years (92 076.23 person-years of follow-up). After adjusting for baseline potential confounders, odds of T2DM were greater for participants with the increasing, U-shape, bell-shape, and other shape change vs decreasing change (adjusted OR [aOR] 2.01, 95% CI 1.42-2.81; 1.56, 1.15-2.13; 1.60, 1.17-2.20; and 1.49, 1.13-2.00, respectively). The results were robust in the sensitivity analyses on excluding baseline participants with T2DM. Moreover, the associations remained significant with male sex, age <60 years and body mass index <24 kg/m2 . CONCLUSIONS This retrospective study revealed increased probability of T2DM with increasing, U-shape, bell-shape, and other-shape TG/HDL-C ratio change trajectories, especially with male sex, age <60 years and body mass index <24 kg/m2 .
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Pei Qin
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Yanmei Lou
- Department of Health Management, Beijing Xiaotangshan Hospital, Beijing, People's Republic of China
| | - Ping Zhao
- Department of Health Management, Beijing Xiaotangshan Hospital, Beijing, People's Republic of China
| | - Xue Li
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Ranran Qie
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xiaoyan Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Minghui Han
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Shengbing Huang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Dechen Liu
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yuying Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Yang Li
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Xingjin Yang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yifei Feng
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Changyi Wang
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Jianping Ma
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Xiaolin Peng
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Hongen Chen
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Dan Zhao
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Shan Xu
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Li Wang
- Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, Shenzhen, People's Republic of China
| | - Xinping Luo
- School of Basic Medicine, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Ming Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Dongsheng Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Fulan Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| |
Collapse
|
36
|
FATP2-targeted therapies - A role beyond fatty liver disease. Pharmacol Res 2020; 161:105228. [PMID: 33027714 DOI: 10.1016/j.phrs.2020.105228] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/26/2020] [Accepted: 09/27/2020] [Indexed: 12/31/2022]
Abstract
Fatty acid transport protein 2 (FATP2) is a multifunctional protein whose specific function is determined by the type of located cell, its intracellular location, or organelle-specific interactions. In the different diseases setting, a newfound appreciation for the biological function of FATP2 has come into view. Two main functions of FATP2 are to activate long-chain fatty acids (LCFAs) as a very long-chain acyl-coenzyme A (CoA) synthetase (ACSVL) and to transport LCFAs as a fatty acid transporter. FATP2 is not only involved in the occurrence of nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM), but also plays an important role in lithogenic diet-induced cholelithiasis, the formation of cancer tumor immunity, the progression of chronic kidney disease (CKD), and the regulation of zoledronate-induced nephrotoxicity. Herein, we review the updated information on the role of FATP2 in related diseases. In particular, we discuss the new functions of FATP2 and propose that FATP2 is a potential clinical biomarker and therapeutic target. In conclusion, regulatory strategies for FATP2 may bring new treatment options for cancer and lipid metabolism-related disorders.
Collapse
|
37
|
Chavira-Suárez E, Rosel-Pech C, Polo-Oteyza E, Ancira-Moreno M, Ibarra-González I, Vela-Amieva M, Meraz-Cruz N, Aguilar-Salinas C, Vadillo-Ortega F. Simultaneous evaluation of metabolomic and inflammatory biomarkers in children with different body mass index (BMI) and waist-to-height ratio (WHtR). PLoS One 2020; 15:e0237917. [PMID: 32834003 PMCID: PMC7446833 DOI: 10.1371/journal.pone.0237917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/06/2020] [Indexed: 11/19/2022] Open
Abstract
Metabolic disturbances and systemic pro-inflammatory changes have been reported in children with obesity. However, it is unclear the time-sequence of metabolic or inflammatory modifications during children obesity evolution. Our study aimed to quantify simultaneously metabolomic and inflammatory biomarkers in serum from children with different levels of adiposity. For this purpose, a cross-sectional study was used to perform targeted metabolomics and inflammatory cytokines measurements. Serum samples from children between six to ten years old were analyzed using either body mass index (BMI) or waist-to-height ratio (WHtR) classifications. One hundred and sixty-eight school-aged children were included. BMI classification in children with overweight or obesity showed altered concentrations of glucose and amino acids (glycine and tyrosine). Children classified by WHtR exhibited imbalances in amino acids (glycine, valine, and tyrosine) and lipids (triacyl glycerides and low-density lipoprotein) compared to control group. No differences in systemic inflammation biomarkers or in the prevalence of other results were found in these children. Abnormal arterial blood pressure was found in 32% of children with increased adiposity. In conclusion, obesity in school-aged children is characterized by significant metabolic modifications that are not accompanied by major disturbances in circulating concentrations of inflammatory biomarkers.
Collapse
Affiliation(s)
- Erika Chavira-Suárez
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Cecilia Rosel-Pech
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología “Dr. Daniel Méndez Hernández" Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Ernestina Polo-Oteyza
- Fondo Nestlé para la Nutrición, Fundación Mexicana Para la Salud, Mexico City, Mexico
| | - Mónica Ancira-Moreno
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Health Department, Universidad Iberoamericana, A.C., Mexico City, Mexico
| | - Isabel Ibarra-González
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | - Noemi Meraz-Cruz
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Carlos Aguilar-Salinas
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Felipe Vadillo-Ortega
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Dirección de Investigación, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
38
|
Manieri E, Folgueira C, Rodríguez ME, Leiva-Vega L, Esteban-Lafuente L, Chen C, Cubero FJ, Barrett T, Cavanagh-Kyros J, Seruggia D, Rosell A, Sanchez-Cabo F, Gómez MJ, Monte MJ, G Marin JJ, Davis RJ, Mora A, Sabio G. JNK-mediated disruption of bile acid homeostasis promotes intrahepatic cholangiocarcinoma. Proc Natl Acad Sci U S A 2020; 117:16492-16499. [PMID: 32601222 PMCID: PMC7368313 DOI: 10.1073/pnas.2002672117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Metabolic stress causes activation of the cJun NH2-terminal kinase (JNK) signal transduction pathway. It is established that one consequence of JNK activation is the development of insulin resistance and hepatic steatosis through inhibition of the transcription factor PPARα. Indeed, JNK1/2 deficiency in hepatocytes protects against the development of steatosis, suggesting that JNK inhibition represents a possible treatment for this disease. However, the long-term consequences of JNK inhibition have not been evaluated. Here we demonstrate that hepatic JNK controls bile acid production. We found that hepatic JNK deficiency alters cholesterol metabolism and bile acid synthesis, conjugation, and transport, resulting in cholestasis, increased cholangiocyte proliferation, and intrahepatic cholangiocarcinoma. Gene ablation studies confirmed that PPARα mediated these effects of JNK in hepatocytes. This analysis highlights potential consequences of long-term use of JNK inhibitors for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Elisa Manieri
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - María Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Laura Esteban-Lafuente
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Chaobo Chen
- Department of Immunology, Ophthalmology, and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology, and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Tamera Barrett
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Julie Cavanagh-Kyros
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Davide Seruggia
- Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| | - Alejandro Rosell
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Fátima Sanchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Manuel Jose Gómez
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Maria J Monte
- Laboratory of Experimental Hepatology and Drug Targeting, National Institute for Study of Liver and Gastrointestinal Diseases (CIBERehd), University of Salamanca, 37007 Salamanca, Spain
| | - Jose J G Marin
- Laboratory of Experimental Hepatology and Drug Targeting, National Institute for Study of Liver and Gastrointestinal Diseases (CIBERehd), University of Salamanca, 37007 Salamanca, Spain
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605;
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain;
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain;
| |
Collapse
|
39
|
Jang YS, Kim HY, Zuo G, Lee EH, Kang SK, Lim SS. Constituents from Solidago virgaurea var. gigantea and their inhibitory effect on lipid accumulation. Fitoterapia 2020; 146:104683. [PMID: 32634454 DOI: 10.1016/j.fitote.2020.104683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022]
Abstract
In this study, the anti-adipogenic activities of compounds isolated from Solidago viraurea var. gigantea (SG) extracts were investigated using Oil Red O staining in the 3T3-L1 cell line. Four known compounds including 3,5-di-O-caffeoylquinic acid (5), protocatechuic acid (6), chlorogenic acid (7), and kaempferol-3-O-rutinoside (8), and four undescribed compounds including (1R,2S,3S,5R,7S)-methyl 7-((cinnamoyloxy)methyl)-2,3-dihydroxy-6,8-dioxabicyclo[3.2.1]octane-5-carboxylate (1), (1R,2S,3S,5R,7S)-methyl 2,3-dihydroxy-7-((((Z)-3-phenylacryloyl)oxy)methyl)-6,8-dioxabicyclo[3.2.1]octane-5-carboxylate (2), (1R,2S,3S,5R,7S)-2,3-dihydroxy-7-((((Z)-3-phenylacryloyl)oxy)methyl)-6,8-dioxabicyclo[3.2.1]octane-5-carboxylic acid (3), and (1R,2S,3S,5R,7S)-7-((cinnamoyloxy)methyl)-2,3-dihydroxy-6,8-dioxabicyclo[3.2.1]octane-5-carboxylic acid (4) were isolated from S. viraurea var. gigantea. The structures of the compounds were first identified by comparing their 1H NMR spectra with spectral data from the literature and a more detailed identification was then performed using 2D NMR (Correlated spectroscopy (COSY), heteronuclear single quantum correlation (HSQC), heteronuclear multiple bond correlation (HMBC), and nuclear overhauser spectroscopy (NOESY)), and X-ray crystallography analyses. The anti-adipogenic activities of all compounds were evaluated by MTT assay and Oil Red O staining in 3T3-L1 cells. 3,5-di-O-caffeoylquinic acid was found to inhibit lipid accumulation more potently than the other tested compounds.
Collapse
Affiliation(s)
- Young Soo Jang
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Republic of Korea
| | - Hyun-Yong Kim
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Republic of Korea
| | - Guanglei Zuo
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Republic of Korea
| | - Eun Hee Lee
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 339-700, Republic of Korea
| | - Sung Kwon Kang
- Department of Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-746, Republic of Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Republic of Korea; Institute of Korean Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Republic of Korea; Institute of Natural Medicine, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Republic of Korea.
| |
Collapse
|
40
|
Zhang D, Cheng C, Cao M, Wang T, Chen X, Zhao Y, Wang B, Ren Y, Liu D, Liu L, Chen X, Liu F, Zhou Q, Tian G, Li Q, Guo C, Li H, Wang J, Cheng R, Hu D, Zhang M. TXNIP hypomethylation and its interaction with obesity and hypertriglyceridemia increase type 2 diabetes mellitus risk: A nested case-control study. J Diabetes 2020; 12:512-520. [PMID: 31919985 DOI: 10.1111/1753-0407.13021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/15/2019] [Accepted: 01/01/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This study aims to estimate type 2 diabetes mellitus (T2DM) incidence with DNA methylation of the thioredoxin-interacting protein (TXNIP) gene and its interaction with environmental factors. MATERIALS AND METHODS This case-control study included 286 incident T2DM cases and 286 non-T2DM controls matched by sex, age, marital status, race, and residence village nested in the Rural Chinese Cohort Study. A conditional logistic regression model was used to estimate the association of DNA methylation at TXNIP gene with T2DM risk. Also, multifactor dimensionality reduction (MDR) and classification and regression tree (CART) analyses were used to investigate the interaction between TXNIP methylation and environmental risk factors. RESULTS Methylation levels of all five CpG loci at TXNIP gene were significantly lower in T2DM than in controls (all P < .001). With increasing methylation level, risk of T2DM was significantly decreased (odds ratio, 95% CI 0.80, 0.69-0.94 for CpG1; 0.80, 0.69-0.93 for CpG2; 0.70, 0.56-0.88 for CpG3; 0.78, 0.66-0.92 for CpG4; and 0.76, 0.60-0.97 for CpG5). Additionally, the essential interactions among TXNIP methylation, obesity, and hypertriglyceridemia were identified by CART and MDR analyses. On logistic regression analysis, the risk of T2DM was reduced with terminal node 5 (CpG3 methylation ≥72%, nonobesity, normal triglyceride (TG) level, and CpG4 methylation ≥83%) vs terminal node 1 (CpG3 methylation <72%) (odds ratio 95% CI 0.20, 0.10-0.40). CONCLUSIONS TXNIP methylation is associated with T2DM incidence in a Chinese population. Interaction between TXNIP methylation and environmental factors may influence T2DM risk and needs more investigation.
Collapse
Affiliation(s)
- Dongdong Zhang
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Cheng Cheng
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Meng Cao
- Department of Environmental Health, Jinan Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Tieqiang Wang
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Xiaoliang Chen
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Yang Zhao
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Bingyuan Wang
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yongcheng Ren
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Dechen Liu
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Leilei Liu
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Xu Chen
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Feiyan Liu
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Qionggui Zhou
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Gang Tian
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Quanman Li
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Chunmei Guo
- Key Lab of Epidemiology, Department of Infectious Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Honghui Li
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Jian Wang
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Ruirong Cheng
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Dongsheng Hu
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ming Zhang
- Center for Community Health Management, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| |
Collapse
|
41
|
Lim TK, Lee HS, Lee YJ. Triglyceride to HDL-cholesterol ratio and the incidence risk of type 2 diabetes in community dwelling adults: A longitudinal 12-year analysis of the Korean Genome and Epidemiology Study. Diabetes Res Clin Pract 2020; 163:108150. [PMID: 32305400 DOI: 10.1016/j.diabres.2020.108150] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
Abstract
AIMS Serum triglyceride to high-density lipoprotein cholesterol (TG/HDL-C) ratio is known to be associated with cardiometabolic diseases. This study is aimed to evaluate the association between the TG/HDL-C ratio and incident type 2 diabetes with a large-sample, community-based Korean cohort over 12 years. METHODS Among 10,038 participants, a total of 8655 participants aged 40 to 69 years without diabetes were selected from the Korean Genome and Epidemiology Study (KoGES). The baseline TG/HDL-C ratio was divided into quartiles. Newly developed type 2 diabetes was defined by any of the following: a fasting plasma glucose level ≥ 126 mg/dL; a glucose level ≥ 200 mg/dL 2-hours after a 75 g oral glucose tolerance test; an HbA1c ≥ 6.5%; or treatment with anti-diabetic therapy. The hazard ratios (HRs) with 95% confidence intervals (CIs) for incident type 2 diabetes were calculated using multivariate Cox proportional hazards regression models after adjusting for potentially confounding variables. RESULTS During the 12-year follow-up period, type 2 diabetes developed in 1437 subjects (16.6%, 1437/8655), with incidence rate of 2.8-5.0 (over 2 years). Compared to the reference first quartile, the HRs (95% CIs) of incident type 2 diabetes in the second, third, and fourth quartiles increased in a dose-response manner after adjusting for potentially confounding variables. CONCLUSIONS High TG/HDL-C ratio at baseline may be a useful surrogate indicator of future incident type 2 diabetes.
Collapse
Affiliation(s)
- Tae-Kyeong Lim
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Department of Research Affairs, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Jae Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Moon JS, Karunakaran U, Suma E, Chung SM, Won KC. The Role of CD36 in Type 2 Diabetes Mellitus: β-Cell Dysfunction and Beyond. Diabetes Metab J 2020; 44:222-233. [PMID: 32347024 PMCID: PMC7188969 DOI: 10.4093/dmj.2020.0053] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 03/25/2020] [Indexed: 12/24/2022] Open
Abstract
Impaired β-cell function is the key pathophysiology of type 2 diabetes mellitus, and chronic exposure of nutrient excess could lead to this tragedy. For preserving β-cell function, it is essential to understand the cause and mechanisms about the progression of β-cells failure. Glucotoxicity, lipotoxicity, and glucolipotoxicity have been suggested to be a major cause of β-cell dysfunction for decades, but not yet fully understood. Fatty acid translocase cluster determinant 36 (CD36), which is part of the free fatty acid (FFA) transporter system, has been identified in several tissues such as muscle, liver, and insulin-producing cells. Several studies have reported that induction of CD36 increases uptake of FFA in several cells, suggesting the functional interplay between glucose and FFA in terms of insulin secretion and oxidative metabolism. However, we do not currently know the regulating mechanism and physiological role of CD36 on glucolipotoxicity in pancreatic β-cells. Also, the downstream and upstream targets of CD36 related signaling have not been defined. In the present review, we will focus on the expression and function of CD36 related signaling in the pancreatic β-cells in response to hyperglycemia and hyperlipidemia (ceramide) along with the clinical studies on the association between CD36 and metabolic disorders.
Collapse
Affiliation(s)
- Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | | | - Elumalai Suma
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Seung Min Chung
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyu Chang Won
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea.
| |
Collapse
|
43
|
Kotzé-Hörstmann LM, Sadie-Van Gijsen H. Modulation of Glucose Metabolism by Leaf Tea Constituents: A Systematic Review of Recent Clinical and Pre-clinical Findings. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:2973-3005. [PMID: 32105058 DOI: 10.1021/acs.jafc.9b07852] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Leaf teas are widely used as a purported treatment for dysregulated glucose homeostasis. The objective of this study was to systematically evaluate the clinical and cellular-metabolic evidence, published between January 2013 and May 2019, and indexed on PubMed, ScienceDirect, and Web of Science, supporting the use of leaf teas for this purpose. Fourteen randomized controlled trials (RCTs) (13 on Camellia sinensis teas) were included, with mixed results, and providing scant mechanistic information. In contrast, 74 animal and cell culture studies focusing on the pancreas, liver, muscle, and adipose tissue yielded mostly positive results and highlighted enhanced insulin signaling as a recurring target associated with the effects of teas on glucose metabolism. We conclude that more studies, including RCTs and pre-clinical studies examining teas from a wider variety of species beyond C. sinensis, are required to establish a stronger evidence base on the use of leaf teas to normalize glucose metabolism.
Collapse
Affiliation(s)
- Liske M Kotzé-Hörstmann
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Parow 7505, South Africa
| | - Hanél Sadie-Van Gijsen
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Parow 7505, South Africa
| |
Collapse
|
44
|
Liang J, Yi X, Xue M, Chen X, Huang X, Sun Q, Wang T, Zhao C, Yang Y, Gao J, Zhou J, Fan J, Yu M. A retrospective cohort study of preoperative lipid indices and their impact on new-onset diabetes after liver transplantation. J Clin Lab Anal 2020; 34:e23192. [PMID: 31981248 PMCID: PMC7246365 DOI: 10.1002/jcla.23192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 11/17/2019] [Accepted: 12/11/2019] [Indexed: 01/06/2023] Open
Abstract
Background The correlation between preoperative lipid profiles and new‐onset diabetes after transplantation (NODAT) remains relatively unexplored in liver transplant recipients (LTRs). Thus, we aimed to investigate the preoperative lipid profiles in Chinese LTRs and evaluate the different influences of preoperative total cholesterol, total triglycerides (TG), high‐density lipoprotein cholesterol, and low‐density lipoprotein cholesterol on the development of NODAT in both sexes. Methods A total of 767 Chinese LTRs from Zhongshan Hospital were retrospectively evaluated. NODAT was defined according to the American Diabetes Association guidelines; the relationship between each preoperative lipid index and NODAT development was analyzed separately in men and women. Results Pretransplant hypotriglyceridemia was observed in 35.72% of the total LTRs. In men, only the preoperative TG level was significantly associated with incident NODAT after adjusting for potential confounders (hazard ratio 1.37, 95% confidence interval 1.13‐1.66, P = .001). There was a nonlinear relationship between the preoperative TG level and NODAT risk. The risk of NODAT significantly increased with preoperative a TG level above 0.54 mmol/L (log‐likelihood ratio test, P = .043). In women, no significant association was observed. Conclusion Among male LTRs, a higher preoperative TG level, even at a low level within the normal range, was significantly and nonlinearly associated with an increased risk of NODAT.
Collapse
Affiliation(s)
- Jing Liang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xilu Yi
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Endocrinology and Metabolism, Central Hospital of Songjiang District, Shanghai, China
| | - Mengjuan Xue
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Geriatrics and Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Xianying Chen
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Endocrinology and Metabolism, Hainan Provincial Nong Ken Hospital, Hainan, China
| | - Xiaowu Huang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.,Key laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Qiman Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.,Key laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ting Wang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenhe Zhao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yinqiu Yang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Gao
- Center of Clinical Epidemiology and Evidence-based Medicine, Fudan University, Shanghai, China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.,Key laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.,Key laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Mingxiang Yu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
|
46
|
Li B, Qiao L, Yan X, Shi T, Ren D, Zhao Y, Zhao J, Liu W. mRNA expression of genes related to fat deposition during in vitro ovine adipogenesis. CANADIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1139/cjas-2018-0107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Fat deposition in animals involves adipogenic differentiation guided by transcriptional factors and other key factors. To understand the molecular mechanism underlying ovine adipogenic differentiation, the dynamic mRNA expression of key genes related to fat deposition, including peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid-binding protein 4 (FABP4), FABP5, and cellular retinoic acid-binding protein 2 (CRABP2), were analyzed during in vitro differentiation of ovine preadipocytes. The stromal vascular cells from underneath the tail fat tissue of 1-wk-old sheep were isolated and cultured, and the preadipocytes were induced using a cocktail of 3-isobutyl-1-methylxanthine, insulin, dexamethasone, and troglitazone. The cultivated cells were collected at different time points after induced differentiation. The expression levels of PPAR-γ, FABP4, FABP5, and CRABP2 were studied by quantitative real-time polymerase chain reaction. The expressions of these genes in sheep were compared with those in human and mouse retrieved from the Gene Expression Omnibus DataSets. We observed that the expression of PPAR-γ, FABP4, and FABP5 was increased upon differentiation of ovine preadipocytes, as in humans and mice. The expression of CRABP2 was sharply increased from days 0 to 2 after induced differentiation and was subsequently decreased. This expression pattern of CRABP2 was different from that observed in humans and mice. Our results provide new insights into the function of these genes in fat deposition.
Collapse
Affiliation(s)
- Baojun Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Liying Qiao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Xiaoru Yan
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Tao Shi
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Duanyang Ren
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Yanyan Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Junxing Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Wenzhong Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| |
Collapse
|
47
|
Jayasinghe SU, Tankeu AT, Amati F. Reassessing the Role of Diacylglycerols in Insulin Resistance. Trends Endocrinol Metab 2019; 30:618-635. [PMID: 31375395 DOI: 10.1016/j.tem.2019.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022]
Abstract
Skeletal muscle (SM) insulin resistance (IR) plays an important role in the burden of obesity, particularly because it leads to glucose intolerance and type 2 diabetes. Among the mechanisms thought to link IR to obesity is the accumulation, in muscle cells, of different lipid metabolites. Diacylglycerols (DAGs) are subject of particular attention due to reported interactions with the insulin signaling cascade. Given that SM accounts for the majority of insulin-stimulated glucose uptake, this review integrates recent observational and mechanistic works with the sole focus on questioning the role of DAGs in SM IR. Particular attention is given to the subcellular distributions and specific structures of DAGs, highlighting future research directions towards reaching a consensus on the mechanistic role played by DAGs.
Collapse
Affiliation(s)
- Sisitha U Jayasinghe
- Aging and Muscle Metabolism Laboratory, Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Aurel T Tankeu
- Aging and Muscle Metabolism Laboratory, Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Francesca Amati
- Aging and Muscle Metabolism Laboratory, Department of Physiology, University of Lausanne, Lausanne, Switzerland; Institute of Sports Sciences, University of Lausanne, Lausanne, Switzerland; Service of Endocrinology, Diabetology and Metabolism, Department of Medicine, University Hospital and Lausanne University, Lausanne, Switzerland.
| |
Collapse
|
48
|
Lipotoxicity in Kidney, Heart, and Skeletal Muscle Dysfunction. Nutrients 2019; 11:nu11071664. [PMID: 31330812 PMCID: PMC6682887 DOI: 10.3390/nu11071664] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/01/2019] [Accepted: 07/17/2019] [Indexed: 12/19/2022] Open
Abstract
Dyslipidemia is a common nutritional and metabolic disorder in patients with chronic kidney disease. Accumulating evidence supports the hypothesis that prolonged metabolic imbalance of lipids leads to ectopic fat distribution in the peripheral organs (lipotoxicity), including the kidney, heart, and skeletal muscle, which accelerates peripheral inflammation and afflictions. Thus, lipotoxicity may partly explain progression of renal dysfunction and even extrarenal complications, including renal anemia, heart failure, and sarcopenia. Additionally, endoplasmic reticulum stress activated by the unfolded protein response pathway plays a pivotal role in lipotoxicity by modulating the expression of key enzymes in lipid synthesis and oxidation. Here, we review the molecular mechanisms underlying lipid deposition and resultant tissue damage in the kidney, heart, and skeletal muscle, with the goal of illuminating the nutritional aspects of these pathologies.
Collapse
|
49
|
Wang X, Wang QC, Sun Z, Li T, Yang K, An C, Guo C, Tang TS. ER stress mediated degradation of diacylglycerol acyltransferase impairs mitochondrial functions in TMCO1 deficient cells. Biochem Biophys Res Commun 2019; 512:914-920. [DOI: 10.1016/j.bbrc.2019.03.115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/18/2019] [Indexed: 12/17/2022]
|
50
|
Cheng FR, Cui HX, Fang JL, Yuan K, Guo Y. Ameliorative Effect and Mechanism of the Purified Anthraquinone-Glycoside Preparation from Rheum Palmatum L. on Type 2 Diabetes Mellitus. Molecules 2019; 24:E1454. [PMID: 31013790 PMCID: PMC6515271 DOI: 10.3390/molecules24081454] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/27/2019] [Accepted: 04/04/2019] [Indexed: 12/11/2022] Open
Abstract
Rheum palmatum L. is a traditional Chinese medicine with various pharmacological properties, including anti-inflammatory, antibacterial, and detoxification effects. In this study, the mechanism of the hypoglycemic effect of purified anthraquinone-Glycoside from Rheum palmatum L. (PAGR) in streptozotocin (STZ) and high-fat diet induced type 2 diabetes mellitus (T2DM) in rats was investigated. The rats were randomly divided into normal (NC), T2DM, metformin (Met), low, middle (Mid), and high (Hig) does of PAGR groups. After six weeks of continuous administration of PAGR, the serum indices and tissue protein expression were determined, and the pathological changes in liver, kidney, and pancreas tissues were observed. The results showed that compared with the type 2 diabetes mellitus group, the fasting blood glucose (FBG), total cholesterol (TC), and triglyceride (TG) levels in the serum of rats in the PAGR treatment groups were significantly decreased, while superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) levels were noticeably increased. The expression of Fas ligand (FasL), cytochrome C (Cyt-c), and caspase-3 in pancreatic tissue was obviously decreased, and the pathological damage to the liver, kidney, and pancreas was improved. These indicate that PAGR can reduce oxidative stress in rats with diabetes mellitus by improving blood lipid metabolism and enhancing their antioxidant capacity, thereby regulating the mitochondrial apoptotic pathway to inhibitβ-cell apoptosis and improve β-cell function. Furthermore, it can regulate Fas/FasL-mediated apoptosis signaling pathway to inhibit β-cell apoptosis, thereby lowering blood glucose levels and improving T2DM.
Collapse
Affiliation(s)
- Fang-Rong Cheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Hong-Xin Cui
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China.
| | - Ji-Li Fang
- Jiyang College of Zhejiang Agriculture and Forestry University, Zhu'ji 311800, China.
| | - Ke Yuan
- Jiyang College of Zhejiang Agriculture and Forestry University, Zhu'ji 311800, China.
| | - Ying Guo
- Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, China.
| |
Collapse
|