1
|
Cui Y, Xu Z, Cui Z, Guo Y, Wu P, Zhou X. Comparative study of insulin resistance surrogate indices to predict mild cognitive impairment among Chinese non-diabetic adults. Lipids Health Dis 2024; 23:357. [PMID: 39487494 DOI: 10.1186/s12944-024-02353-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
OBJECTIVE The study aims to investigate the associations of triglyceride glucose (TyG), TyG combined with body mass index (TyG-BMI), metabolic score for insulin resistance (METS-IR), the triglyceride to high-density lipoprotein cholesterol ratio (TG/HDL-C), and the risk of mild cognitive impairment (MCI) in individuals without diabetes aged 45 and above. METHODS The most recent data in this study were from a cohort study, which sourced samples from the China Health and Retirement Longitudinal Study, spanning 2011 to 2018. The four indices' associations with MCI risk were analyzed using logistic regression. The predictive capacity was measured using the receiver operating characteristic (ROC) curve. RESULTS Over a 7-year follow-up, 1,261 individuals (31.34%) of the 4,027 participants developed MCI. Logistic regression analysis revealed significant associations between these surrogate indices and MCI. The findings for the highest quartile compared with the lowest quartile were as follows: TyG: 1.24 (95% CI: 1.02, 1.49); TyG-BMI: 1.38 (95% CI: 1.13, 1.68); METS-IR: 1.50 (95% CI: 1.09, 2.06); TG/HDL-C: 1.34 (95% CI: 1.10, 1.64). ROC analysis revealed that TyG, TyG-BMI, TG/HDL-C, and METS-IR demonstrated excellent discriminatory power for MCI, with area under the curve (AUC) values of 0.82 (95% CI: 0.80, 0.83), 0.82 (95% CI: 0.80, 0.83), 0.83 (95% CI: 0.80, 0.84), and 0.83 (95% CI: 0.80, 0.84), respectively. The four indices showed stronger ability to predict MCI risk in females compared to males. CONCLUSION Elevated levels of four indices are positively correlated with MCI risk. TyG-BMI and METS-IR demonstrate stronger capabilities in identifying MCI across both male and female populations. This suggests that early intervention in patients with elevated IR surrogate indices may help reduce the MCI.
Collapse
Affiliation(s)
- Yuyu Cui
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Zhening Xu
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Zhaoshu Cui
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Yuanyuan Guo
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Peiwei Wu
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Xiaoyan Zhou
- School of Medicine, Yan'an University, Yan'an, 716000, China.
| |
Collapse
|
2
|
Wang X, Jiang R, Shen J, Chen S, Wu S, Hu H, Cai H. Transitions in metabolic syndrome and metabolic obesity status over time and risk of urologic cancer: A prospective cohort study. PLoS One 2024; 19:e0311492. [PMID: 39432545 PMCID: PMC11493304 DOI: 10.1371/journal.pone.0311492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/18/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND AND AIMS The effects of metabolic obesity (MO) phenotypes status and their dynamic changes on urologic cancer (UC) is ignored. We aimed to investigate the association between metabolic syndrome (MetS) and MO status at baseline, their dynamic changes and UC risk. METHODS This paper studied 97,897 subjects who were free of cancers at baseline (2006-2007). Individuals were classified into four MO phenotypes by MetS and obesity at baseline. Transitions in MetS and MO status from 2006-2007 to 2008-2009 were considered. The hazard ratios (HRs) and 95% confidence intervals (CIs) for UC were assessed by multifactorial Cox proportional risk regression models. The main limitations of this study are as follows: the ratio of men to women in the cohort is unbalanced; the impacts of MetS and MO on each cancer type (kidney cancer, prostate cancer, bladder cancer) have not been analyzed separately; the transition intervals of MetS and MO phenotypes are relatively short. RESULTS From baseline (2006-2007) survey to December 31, 2020, during a median follow-up of 14.02 years, 554 cases of UC were diagnosed. Participants with MetS [HRs (95% CI) = 1.26 (1.06-1.49)] and metabolically unhealthy obesity (MUO) [HRs (95% CI) = 1.49 (1.17-1.89)] had significantly higher risk of UC than those with non-MetS and metabolically healthy normal weight (MHN). Transitions in MetS and MO phenotypes over time were studied. Compared with non-MetS to non-MetS, the risks for UC in MetS to MetS [HRs (95% CI) = 1.45 (1.11-1.88)] was increased. Compared with MHN to MHN, both MUO to metabolically healthy obesity (MHO) [HRs (95% CI) = 2.65 (1.43-4.92)] and MUO to MUO [HRs (95% CI) = 1.60 (1.06-2.42)] had significantly higher UC risk. CONCLUSIONS MetS and MUO increased the UC risk at baseline. Transitions of MetS to MetS, MUO to MUO and even MUO to MHO over time significantly increased the risk of UC development.
Collapse
Affiliation(s)
- Xia Wang
- Department of Gynaecology, Tangshan Hongci Hospital, Tangshan, Hebei, China
| | - Runxue Jiang
- Department of Oncology Surgery, Tangshan People’s Hospital, Tangshan, Hebei, China
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jianglun Shen
- Department of Oncology Surgery, Tangshan People’s Hospital, Tangshan, Hebei, China
| | - Shuohua Chen
- Health Department of Kailuan(Group), Tangshan, Hebei, China
| | - Shouling Wu
- Health Department of Kailuan(Group), Tangshan, Hebei, China
| | - Hailong Hu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haifeng Cai
- Department of Oncology Surgery, Tangshan People’s Hospital, Tangshan, Hebei, China
| |
Collapse
|
3
|
Sergi D, Melloni M, Passaro A, Neri LM. Influence of Type 2 Diabetes and Adipose Tissue Dysfunction on Breast Cancer and Potential Benefits from Nutraceuticals Inducible in Microalgae. Nutrients 2024; 16:3243. [PMID: 39408212 PMCID: PMC11478231 DOI: 10.3390/nu16193243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Breast cancer (BC) represents the most prevalent cancer in women at any age after puberty. From a pathogenetic prospective, despite a wide array of risk factors being identified thus far, poor metabolic health is emerging as a putative risk factor for BC. In particular, type 2 diabetes mellitus (T2DM) provides a perfect example bridging the gap between poor metabolic health and BC risk. Indeed, T2DM is preceded by a status of hyperinsulinemia and is characterised by hyperglycaemia, with both factors representing potential contributors to BC onset and progression. Additionally, the aberrant secretome of the dysfunctional, hypertrophic adipocytes, typical of obesity, characterised by pro-inflammatory mediators, is a shared pathogenetic factor between T2DM and BC. In this review, we provide an overview on the effects of hyperglycaemia and hyperinsulinemia, hallmarks of type 2 diabetes mellitus, on breast cancer risk, progression, treatment and prognosis. Furthermore, we dissect the role of the adipose-tissue-secreted adipokines as additional players in the pathogenesis of BC. Finally, we focus on microalgae as a novel superfood and a source of nutraceuticals able to mitigate BC risk by improving metabolic health and targeting cellular pathways, which are disrupted in the context of T2DM and obesity.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Luca Maria Neri
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
- Laboratory for Technologies of Advanced Therapies (LTTA)—Electron Microscopy Center, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
4
|
Shao W, Pan B, Li Z, Peng R, Yang W, Xie Y, Han D, Fang X, Li J, Zhu Y, Zhao Z, Kan H, Ying Z, Xu Y. Gut microbiota mediates ambient PM 2.5 exposure-induced abnormal glucose metabolism via short-chain fatty acids. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135096. [PMID: 38996677 PMCID: PMC11342392 DOI: 10.1016/j.jhazmat.2024.135096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
PM2.5 exposure has been found to cause gut dysbiosis and impair glucose homeostasis in human and animals, yet their underlying biological connection remain unclear. In the present study, we aim to investigate the biological significance of gut microbiota in PM2.5-induced glucose metabolic abnormalities. Our results showed that microbiota depletion by antibiotics treatment significantly alleviated PM2.5-induced glucose intolerance and insulin resistance, as indicated by the intraperitoneal glucose tolerance test, glucose-induced insulin secretion, insulin tolerance test, insulin-induced phosphorylation levels of Akt and GSK-3β in insulin sensitive tissues. In addition, faecal microbiota transplantation (FMT) from PM2.5-exposed donor mice successfully remodeled the glucose metabolism abnormalities in recipient mice, while the transplantation of autoclaved faecal materials did not. Faecal microbiota analysis demonstrated that the composition and alpha diversity of the gut bacterial community were altered by PM2.5 exposure and in FMT recipient mice. Furthermore, short-chain fatty acids levels analysis showed that the circulating acetate was significantly decreased in PM2.5-exposed donor and FMT recipient mice, and supplementation of sodium acetate for 3 months successfully improved the glucose metabolism abnormalities induced by PM2.5 exposure. These results indicate that manipulating gut microbiota or its metabolites could be a potential strategy for preventing the adverse health effects of ambient PM2.5.
Collapse
Affiliation(s)
- Wenpu Shao
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Bin Pan
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Zhouzhou Li
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Renzhen Peng
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Wenhui Yang
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Yuanting Xie
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Dongyang Han
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Xinyi Fang
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Jingyu Li
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Yaning Zhu
- Department of Pathology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, China.
| | - Zhuohui Zhao
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Mladenović D, Vesković M, Šutulović N, Hrnčić D, Stanojlović O, Radić L, Macut JB, Macut D. Adipose-derived extracellular vesicles - a novel cross-talk mechanism in insulin resistance, non-alcoholic fatty liver disease, and polycystic ovary syndrome. Endocrine 2024; 85:18-34. [PMID: 38285412 DOI: 10.1007/s12020-024-03702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/14/2024] [Indexed: 01/30/2024]
Abstract
Obesity is the best described risk factor for the development of non-alcoholic fatty liver disease (NAFLD)/metabolic dysfunction associated steatotic liver disease (MASLD) and polycystic ovary syndrome (PCOS) while the major pathogenic mechanism linking these entities is insulin resistance (IR). IR is primarily caused by increased secretion of proinflammatory cytokines, adipokines, and lipids from visceral adipose tissue. Increased fatty acid mobilization results in ectopic fat deposition in the liver which causes endoplasmic reticulum stress, mitochondrial dysfunction, and oxidative stress resulting in increased cytokine production and subsequent inflammation. Similarly, IR with hyperinsulinemia cause hyperandrogenism, the hallmark of PCOS, and inflammation in the ovaries. Proinflammatory cytokines from both liver and ovaries aggravate IR thus providing a complex interaction between adipose tissue, liver, and ovaries in inducing metabolic abnormalities in obese subjects. Although many pathogenic mechanisms of IR, NAFLD/MASLD, and PCOS are known, there is still no effective therapy for these entities suggesting the need for further evaluation of their pathogenesis. Extracellular vesicles (EVs) represent a novel cross-talk mechanism between organs and include membrane-bound vesicles containing proteins, lipids, and nucleic acids that may change the phenotype and function of target cells. Adipose tissue releases EVs that promote IR, the development of all stages of NAFLD/MASLD and PCOS, while mesenchymal stem cell-derived AVs may alleviate metabolic abnormalities and may represent a novel therapeutic device in NAFLD/MASLD, and PCOS. The purpose of this review is to summarize the current knowledge on the role of adipose tissue-derived EVs in the pathogenesis of IR, NAFLD/MASLD, and PCOS.
Collapse
Affiliation(s)
- Dušan Mladenović
- Institute of Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Milena Vesković
- Institute of Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Šutulović
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Hrnčić
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Stanojlović
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Lena Radić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Jelica Bjekić Macut
- University of Belgrade Faculty of Medicine, Department of Endocrinology, UMC Bežanijska kosa, Belgrade, Serbia
| | - Djuro Macut
- University of Belgrade Faculty of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| |
Collapse
|
6
|
Xing Z, Chen H, Alman AC. Discriminating insulin resistance in middle-aged nondiabetic women using machine learning approaches. AIMS Public Health 2024; 11:667-687. [PMID: 39027391 PMCID: PMC11252584 DOI: 10.3934/publichealth.2024034] [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: 02/20/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 07/20/2024] Open
Abstract
Objective We employed machine learning algorithms to discriminate insulin resistance (IR) in middle-aged nondiabetic women. Methods The data was from the National Health and Nutrition Examination Survey (2007-2018). The study subjects were 2084 nondiabetic women aged 45-64. The analysis included 48 predictors. We randomly divided the data into training (n = 1667) and testing (n = 417) datasets. Four machine learning techniques were employed to discriminate IR: extreme gradient boosting (XGBoosting), random forest (RF), gradient boosting machine (GBM), and decision tree (DT). The area under the curve (AUC) of receiver operating characteristic (ROC), accuracy, sensitivity, specificity, positive predictive value, negative predictive value, and F1 score were compared as performance metrics to select the optimal technique. Results The XGBoosting algorithm achieved a relatively high AUC of 0.93 in the training dataset and 0.86 in the testing dataset to discriminate IR using 48 predictors and was followed by the RF, GBM, and DT models. After selecting the top five predictors to build models, the XGBoost algorithm with the AUC of 0.90 (training dataset) and 0.86 (testing dataset) remained the optimal prediction model. The SHapley Additive exPlanations (SHAP) values revealed the associations between the five predictors and IR, namely BMI (strongly positive impact on IR), fasting glucose (strongly positive), HDL-C (medium negative), triglycerides (medium positive), and glycohemoglobin (medium positive). The threshold values for identifying IR were 29 kg/m2, 100 mg/dL, 54.5 mg/dL, 89 mg/dL, and 5.6% for BMI, glucose, HDL-C, triglycerides, and glycohemoglobin, respectively. Conclusion The XGBoosting algorithm demonstrated superior performance metrics for discriminating IR in middle-aged nondiabetic women, with BMI, glucose, HDL-C, glycohemoglobin, and triglycerides as the top five predictors.
Collapse
Affiliation(s)
- Zailing Xing
- College of Public Health, University of South Florida, 13201 Bruce B. Downs Blvd, MDC 56, Tampa, FL 33612, USA
| | | | | |
Collapse
|
7
|
Lu QS, Ma L, Jiang WJ, Wang XB, Lu M. KAT7/HMGN1 signaling epigenetically induces tyrosine phosphorylation-regulated kinase 1A expression to ameliorate insulin resistance in Alzheimer's disease. World J Psychiatry 2024; 14:445-455. [PMID: 38617985 PMCID: PMC11008392 DOI: 10.5498/wjp.v14.i3.445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/31/2023] [Accepted: 02/01/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Epidemiological studies have revealed a correlation between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2D). Insulin resistance in the brain is a common feature in patients with T2D and AD. KAT7 is a histone acetyltransferase that participates in the modulation of various genes. AIM To determine the effects of KAT7 on insulin patients with AD. METHODS APPswe/PS1-dE9 double-transgenic and db/db mice were used to mimic AD and diabetes, respectively. An in vitro model of AD was established by Aβ stimulation. Insulin resistance was induced by chronic stimulation with high insulin levels. The expression of microtubule-associated protein 2 (MAP2) was assessed using immunofluorescence. The protein levels of MAP2, Aβ, dual-specificity tyrosine phosphorylation-regulated kinase-1A (DYRK1A), IRS-1, p-AKT, total AKT, p-GSK3β, total GSK3β, DYRK1A, and KAT7 were measured via western blotting. Accumulation of reactive oxygen species (ROS), malondialdehyde (MDA), and SOD activity was measured to determine cellular oxidative stress. Flow cytometry and CCK-8 assay were performed to evaluate neuronal cell death and proliferation, respectively. Relative RNA levels of KAT7 and DYRK1A were examined using quantitative PCR. A chromatin immunoprecipitation assay was conducted to detect H3K14ac in DYRK1A. RESULTS KAT7 expression was suppressed in the AD mice. Overexpression of KAT7 decreased Aβ accumulation and MAP2 expression in AD brains. KAT7 overexpression decreased ROS and MDA levels, elevated SOD activity in brain tissues and neurons, and simultaneously suppressed neuronal apoptosis. KAT7 upregulated levels of p-AKT and p-GSK3β to alleviate insulin resistance, along with elevated expression of DYRK1A. KAT7 depletion suppressed DYRK1A expression and impaired H3K14ac of DYRK1A. HMGN1 overexpression recovered DYRK1A levels and reversed insulin resistance caused by KAT7 depletion. CONCLUSION We determined that KAT7 overexpression recovered insulin sensitivity in AD by recruiting HMGN1 to enhance DYRK1A acetylation. Our findings suggest that KAT7 is a novel and promising therapeutic target for the resistance in AD.
Collapse
Affiliation(s)
- Qun-Shan Lu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Lin Ma
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Wen-Jing Jiang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xing-Bang Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Mei Lu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
8
|
Yuan W, Lin H, Sun Y, Liu L, Yan M, Song Y, Zhang X, Lu X, Xu Y, He Q, Ouyang K, Zhang C, Pan Y, Huang Y, Li Y, Lu X, Liu J. Myocardin reverses insulin resistance and ameliorates cardiomyopathy by increasing IRS-1 expression in a murine model of lipodystrophy caused by adipose deficiency of vacuolar H +-ATPase V0d1 subunit. Theranostics 2024; 14:2246-2264. [PMID: 38505620 PMCID: PMC10945344 DOI: 10.7150/thno.93192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/02/2024] [Indexed: 03/21/2024] Open
Abstract
Aim: Adipose tissue (AT) dysfunction that occurs in both obesity and lipodystrophy is associated with the development of cardiomyopathy. However, it is unclear how dysfunctional AT induces cardiomyopathy due to limited animal models available. We have identified vacuolar H+-ATPase subunit Vod1, encoded by Atp6v0d1, as a master regulator of adipogenesis, and adipose-specific deletion of Atp6v0d1 (Atp6v0d1AKO) in mice caused generalized lipodystrophy and spontaneous cardiomyopathy. Using this unique animal model, we explore the mechanism(s) underlying lipodystrophy-related cardiomyopathy. Methods and Results: Atp6v0d1AKO mice developed cardiac hypertrophy at 12 weeks, and progressed to heart failure at 28 weeks. The Atp6v0d1AKO mouse hearts exhibited excessive lipid accumulation and altered lipid and glucose metabolism, which are typical for obesity- and diabetes-related cardiomyopathy. The Atp6v0d1AKO mice developed cardiac insulin resistance evidenced by decreased IRS-1/2 expression in hearts. Meanwhile, the expression of forkhead box O1 (FoxO1), a transcription factor which plays critical roles in regulating cardiac lipid and glucose metabolism, was increased. RNA-seq data and molecular biological assays demonstrated reduced expression of myocardin, a transcription coactivator, in Atp6v0d1AKO mouse hearts. RNA interference (RNAi), luciferase reporter and ChIP-qPCR assays revealed the critical role of myocardin in regulating IRS-1 transcription through the CArG-like element in IRS-1 promoter. Reducing IRS-1 expression with RNAi increased FoxO1 expression, while increasing IRS-1 expression reversed myocardin downregulation-induced FoxO1 upregulation in cardiomyocytes. In vivo, restoring myocardin expression specifically in Atp6v0d1AKO cardiomyocytes increased IRS-1, but decreased FoxO1 expression. As a result, the abnormal expressions of metabolic genes in Atp6v0d1AKO hearts were reversed, and cardiac dysfunctions were ameliorated. Myocardin expression was also reduced in high fat diet-induced diabetic cardiomyopathy and palmitic acid-treated cardiomyocytes. Moreover, increasing systemic insulin resistance with rosiglitazone restored cardiac myocardin expression and improved cardiac functions in Atp6v0d1AKO mice. Conclusion: Atp6v0d1AKO mice are a novel animal model for studying lipodystrophy- or metabolic dysfunction-related cardiomyopathy. Moreover, myocardin serves as a key regulator of cardiac insulin sensitivity and metabolic homeostasis, highlighting myocardin as a potential therapeutic target for treating lipodystrophy- and diabetes-related cardiomyopathy.
Collapse
Affiliation(s)
- Wenlin Yuan
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Hui Lin
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yuan Sun
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Pharmacology, College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Lihuan Liu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Meijuan Yan
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yujuan Song
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiaofan Zhang
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiangling Lu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yipei Xu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qiyue He
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chenglin Zhang
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yong Pan
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ying Li
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xifeng Lu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Jie Liu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
9
|
Tsilingiris D, Vallianou NG, Spyrou N, Kounatidis D, Christodoulatos GS, Karampela I, Dalamaga M. Obesity and Leukemia: Biological Mechanisms, Perspectives, and Challenges. Curr Obes Rep 2024; 13:1-34. [PMID: 38159164 PMCID: PMC10933194 DOI: 10.1007/s13679-023-00542-z] [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] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW To examine the epidemiological data on obesity and leukemia; evaluate the effect of obesity on leukemia outcomes in childhood acute lymphoblastic leukemia (ALL) survivors; assess the potential mechanisms through which obesity may increase the risk of leukemia; and provide the effects of obesity management on leukemia. Preventive (diet, physical exercise, obesity pharmacotherapy, bariatric surgery) measures, repurposing drugs, candidate therapeutic agents targeting oncogenic pathways of obesity and insulin resistance in leukemia as well as challenges of the COVID-19 pandemic are also discussed. RECENT FINDINGS Obesity has been implicated in the development of 13 cancers, such as breast, endometrial, colon, renal, esophageal cancers, and multiple myeloma. Leukemia is estimated to account for approximately 2.5% and 3.1% of all new cancer incidence and mortality, respectively, while it represents the most frequent cancer in children younger than 5 years. Current evidence indicates that obesity may have an impact on the risk of leukemia. Increased birthweight may be associated with the development of childhood leukemia. Obesity is also associated with worse outcomes and increased mortality in leukemic patients. However, there are several limitations and challenges in meta-analyses and epidemiological studies. In addition, weight gain may occur in a substantial number of childhood ALL survivors while the majority of studies have documented an increased risk of relapse and mortality among patients with childhood ALL and obesity. The main pathophysiological pathways linking obesity to leukemia include bone marrow adipose tissue; hormones such as insulin and the insulin-like growth factor system as well as sex hormones; pro-inflammatory cytokines, such as IL-6 and TNF-α; adipocytokines, such as adiponectin, leptin, resistin, and visfatin; dyslipidemia and lipid signaling; chronic low-grade inflammation and oxidative stress; and other emerging mechanisms. Obesity represents a risk factor for leukemia, being among the only known risk factors that could be prevented or modified through weight loss, healthy diet, and physical exercise. Pharmacological interventions, repurposing drugs used for cardiometabolic comorbidities, and bariatric surgery may be recommended for leukemia and obesity-related cancer prevention.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100, Alexandroupolis, Greece
| | - Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Nikolaos Spyrou
- Tisch Cancer Institute Icahn School of Medicine at Mount Sinai, 1190 One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Dimitris Kounatidis
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Str, 12462, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, 11527, Athens, Greece.
| |
Collapse
|
10
|
Meng FD, Yuan L, Xu DJ, Che MY, Hou SZ, Lu DD, Liu WJ, Nan Y. Exploring the targets and molecular mechanism of glycyrrhetinic acid against diabetic nephropathy based on network pharmacology and molecular docking. World J Diabetes 2023; 14:1672-1692. [DOI: 10.4239/wjd.v14.i11.1672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/25/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) stands as the most prevalent chronic microvascular complication of diabetes mellitus. Approximately 50% of DN patients progress to end-stage renal disease, posing a substantial health burden.
AIM To employ network pharmacology and molecular docking methods to predict the mechanism by which glycyrrhetinic acid (GA) treats DN, subsequently validating these predictions through experimental means.
METHODS The study initially identified GA targets using Pharm Mapper and the TCMSP database. Targets relevant to DN were obtained from the Genecards, OMIM, and TTD databases. The Venny database facilitated the acquisition of intersecting targets between GA and DN. The String database was used to construct a protein interaction network, while DAVID database was used to conducted Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) analysis. Molecular docking experiments were performed using Autodock software with selected proteins. Experimental validation was conducted using renal proximal tubular cells (HK-2) as the study subjects. A hyperglycemic environment was simulated using glucose solution, and the effect of GA on cell viability was assessed through the cell counting kit-8 method. Flow cytometry was employed to detect cell cycle and apoptosis, and protein immunoblot (western blot) was used to measure the expression of proteins of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway and insulin resistance pathway, including insulin receptor (INSR), PI3K, p-PI3K, AKT, p-AKT, and glycogen synthase kinase-3 (GSK3).
RESULTS A total of 186 intersecting targets between GA and DN were identified, which were associated with 144 KEGG-related enrichment pathways, 375 GO biological process entries, 45 GO cellular component entries, and 112 GO cellular function entries. Molecular docking demonstrated strong binding of GA to mitogen-activated protein kinase (MAPK)-1, SRC, PIK3R1, HSP90AA1, CASPASE9, HARS, KRAS, and MAPK14. In vitro experiments revealed that GA inhibited HK-2 cell viability, induced cell cycle arrest at the G2/M phase, and reduced apoptosis with increasing drug concentration. Western blot analysis showed that GA differentially up-regulated GSK3 protein expression, up-regulated AKT/p-AKT expression, down-regulated INSR, AKT, p-AKT, PI3K, and p-PI3K protein expression, and reduced p-PI3K/PI3K levels under high glucose conditions.
CONCLUSION GA may protect renal intrinsic cells by modulating the PI3K/AKT signaling pathway, thereby inhibiting HK-2 cell viability, reducing HK-2 cell apoptosis, and inducing cell cycle arrest at the G0/G1 phase.
Collapse
Affiliation(s)
- Fan-Di Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Duo-Jie Xu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Meng-Ying Che
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shao-Zhang Hou
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Wen-Jing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
11
|
Cortez NE, Lanzi CR, Vahmani P, Matsukuma K, Mackenzie GG. Hepatic safety profile of pancreatic cancer‑bearing mice fed a ketogenic diet in combination with gemcitabine. Oncol Lett 2023; 26:479. [PMID: 37818128 PMCID: PMC10561147 DOI: 10.3892/ol.2023.14067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Ketogenic diets (KDs) are actively being evaluated for their potential anticancer effects. Although KDs are generally considered safe, their safety profile when combined with chemotherapy remains unknown. It is known that a KD enhances the anticancer effect of gemcitabine (2',2'-difluoro-2'-deoxycytidine) in LSL-KrasLSL-G12D/+Trp53R172H/+Pdx-1-Cre (KPC) tumor-bearing mice. In the present study, whether a KD in combination with gemcitabine affected the liver safety profile in KPC mice was evaluated. For this purpose, male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD; % kcal: 20% fat, 65% carbohydrate, 15% protein) + gemcitabine [control plus gemcitabine group (CG)] or a KD (% kcal: 84% fat, 15% protein, 1% carbohydrate) + gemcitabine [ketogenic plus gemcitabine group (KG)] for two months. After two months of treatment, no significant differences in body weight were observed between CGs and KGs. Moreover, the KD did not significantly alter the serum protein expression levels of liver enzymes, including aspartate aminotransferase, alanine aminotransferase and alkaline phosphatase. In addition, the KD did not alter markers of liver-lipid accumulation as well as serum cholesterol and triglyceride levels, compared with the CG-treated group. Upon histologic examination, steatosis was rare, with no notable differences between treatment groups. When examining liver fatty acid composition, KD treatment significantly increased the content of saturated fatty acids and significantly decreased levels of cis-monounsaturated fatty acids compared with the CG. Finally, the KD did not affect liver markers of inflammation and oxidative stress, nor the protein expression levels of enzymes involved in ketone bodies, such as 3-hydroxy-3-methylglutaryl-CoA lyase and hidroximetilglutaril-CoA sintasa, and glucose metabolism, such as hexokinase 2, pyruvate dehydrogenase and phosphofructokinase. In summary, a KD in combination with gemcitabine appears to be safe, with no apparent hepatotoxicity and these data support the further evaluation of a KD as an adjuvant dietary treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Natalia E. Cortez
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | | | - Payam Vahmani
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | - Karen Matsukuma
- Department of Pathology and Laboratory Medicine, Davis Medical Center, University of California, Sacramento, CA 95817, USA
- University of California Davis Comprehensive Cancer Center, University of California, Sacramento, CA 95817, USA
| | - Gerardo G. Mackenzie
- Department of Nutrition, University of California, Davis, CA 95616, USA
- University of California Davis Comprehensive Cancer Center, University of California, Sacramento, CA 95817, USA
| |
Collapse
|
12
|
Metin Z, Tur K, Durmaz K, Akogul S, Akca HM, Imren IG, Demir NBO, Ozkoca D. A comprehensive investigation of novel and traditional inflammatory and metabolic markers as predictive indicators in psoriasis. Int J Dermatol 2023; 62:1272-1280. [PMID: 37610067 DOI: 10.1111/ijd.16813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory and papulosquamous dermatological disorder. While previous studies have discussed certain inflammatory markers for diagnosing and monitoring psoriasis, there is an absence of comprehensive research encompassing both novel and traditional inflammatory markers, as well as metabolic markers, in relation to psoriasis. METHODS A total of 209 individuals participated, including 54 psoriasis patients and 155 controls. Psoriasis Area Severity Index (PASI) was calculated for the patient group. Potential predictive markers for psoriasis were identified: Uric acid/HDL ratio (UHR), D-dimer/albumin ratio (DAR), fibrinogen/albumin ratio (FAR), erythrocyte sedimentation rate, CRP, WBC, HOMA-IR, and vitamin D levels. Differences between groups and correlations with PASI and each other were analyzed using the Mann-Whitney U test and Spearman correlation coefficient. RESULTS The results indicate that the patient group exhibited statistically significantly higher levels of UHR, FAR, CRP, WBC, and HOMA-IR. Upon analyzing the correlations between PASI and the identified markers, statistically significant positive correlation with WBC and negative correlation with vitamin D were observed. The correlations of PASI with other markers did not reach statistical significance. It should be underlined that our study was conducted in a predominantly mild-to-moderate patient population. CONCLUSIONS The absence of specificity of these markers for psoriasis limits their practical application. However, the development of new objective measures by using them in combination with specific data such as PASI will provide significant benefits in terms of disease diagnosis, follow-up, and treatment.
Collapse
Affiliation(s)
- Zuhal Metin
- Department of Dermatology, Faculty of Medicine, Kirsehir Ahi Evran University, Kirsehir, Turkey
| | - Kagan Tur
- Department of Internal Medicine, Faculty of Medicine, Kirsehir Ahi Evran University, Kirsehir, Turkey
| | - Koray Durmaz
- Department of Dermatology, Lokman Hekim Etlik Hospital, Ankara, Turkey
| | - Serkan Akogul
- Department of Statistics, Faculty of Sciences, Pamukkale University, Denizli, Turkey
| | - Hanife M Akca
- Department of Dermatology, Faculty of Medicine, Karamanoglu Mehmet Bey University, Karaman, Turkey
| | - Işıl G Imren
- Department of Dermatology, Denizli State Hospital, Denizli, Turkey
| | - Nazime B O Demir
- Department of Dermatology, Faculty of Medicine, Kirsehir Ahi Evran University, Kirsehir, Turkey
| | - Defne Ozkoca
- Department of Dermatology, Zonguldak Ataturk State Hospital, Zonguldak, Turkey
| |
Collapse
|
13
|
Hopland-Nechita FV, Andersen JR, Rajalahti TK, Andreassen T, Beisland C. Identifying possible biomarkers of lower urinary tract symptoms using metabolomics and partial least square regression. Metabolomics 2023; 19:82. [PMID: 37698748 PMCID: PMC10497431 DOI: 10.1007/s11306-023-02046-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023]
Abstract
INTRODUCTION The objective of this study was to explore potential novel biomarkers for moderate to severe lower urinary tract symptoms (LUTS) using a metabolomics-based approach, and statistical methods with significant different features than previous reported. MATERIALS AND METHODS The patients and the controls were selected to participate in the study according to inclusion/exclusion criteria (n = 82). We recorded the following variables: International prostatic symptom score (IPSS), prostate volume, comorbidities, PSA, height, weight, triglycerides, glycemia, HDL cholesterol, and blood pressure. The study of 41 plasma metabolites was done using the nuclear magnetic resonance spectroscopy technique. First, the correlations between the metabolites and the IPSS were done using Pearson. Second, significant biomarkers of LUTS from metabolites were further analysed using a multiple linear regression model. Finally, we validated the findings using partial least square regression (PLS). RESULTS Small to moderate correlations were found between IPSS and methionine (-0.301), threonine (-0.320), lactic acid (0.294), pyruvic acid (0.207) and 2-aminobutyric-acid (0.229). The multiple linear regression model revealed that only threonine (p = 0.022) was significantly associated with IPSS, whereas methionine (p = 0.103), lactic acid (p = 0.093), pyruvic acid (p = 0.847) and 2-aminobutyric-acid (p = 0.244) lost their significance. However, all metabolites lost their significance in the PLS model. CONCLUSION When using the robust PLS-regression method, none of the metabolites in our analysis had a significant association with lower urinary tract symptoms. This highlights the importance of using appropriate statistical methods when exploring new biomarkers in urology.
Collapse
Affiliation(s)
- Florin V Hopland-Nechita
- Department of Urology, Førde Central Hospital, Førde, Norway.
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - John R Andersen
- Western Norway University of Applied Sciences, Førde, Norway
| | | | - Trygve Andreassen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Central staff, St. Olavs Hospital HF, Trondheim, Norway
| | - Christian Beisland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Urology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
14
|
Chen Y, Wang W, Morgan MP, Robson T, Annett S. Obesity, non-alcoholic fatty liver disease and hepatocellular carcinoma: current status and therapeutic targets. Front Endocrinol (Lausanne) 2023; 14:1148934. [PMID: 37361533 PMCID: PMC10286797 DOI: 10.3389/fendo.2023.1148934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Obesity is a global epidemic and overwhelming evidence indicates that it is a risk factor for numerous cancers, including hepatocellular carcinoma (HCC), the third leading cause of cancer-related deaths worldwide. Obesity-associated hepatic tumorigenesis develops from nonalcoholic fatty liver disease (NAFLD), progressing to nonalcoholic steatohepatitis (NASH), cirrhosis and ultimately to HCC. The rising incidence of obesity is resulting in an increased prevalence of NAFLD and NASH, and subsequently HCC. Obesity represents an increasingly important underlying etiology of HCC, in particular as the other leading causes of HCC such as hepatitis infection, are declining due to effective treatments and vaccines. In this review, we provide a comprehensive overview of the molecular mechanisms and cellular signaling pathways involved in the pathogenesis of obesity-associated HCC. We summarize the preclinical experimental animal models available to study the features of NAFLD/NASH/HCC, and the non-invasive methods to diagnose NAFLD, NASH and early-stage HCC. Finally, since HCC is an aggressive tumor with a 5-year survival of less than 20%, we will also discuss novel therapeutic targets for obesity-associated HCC and ongoing clinical trials.
Collapse
Affiliation(s)
- Yinshuang Chen
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Maria P. Morgan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Stephanie Annett
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
15
|
Farhadi-Azar M, Noroozzadeh M, Ghahremani M, Rahmati M, Saei Ghare Naz M, Azizi F, Ramezani Tehrani F. Maternal androgen excess increases the risk of pre-diabetes mellitus in male offspring in later life: a long-term population-based follow-up study. J Endocrinol Invest 2023:10.1007/s40618-022-01972-7. [PMID: 37081228 DOI: 10.1007/s40618-022-01972-7] [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: 08/24/2022] [Accepted: 11/20/2022] [Indexed: 04/22/2023]
Abstract
PURPOSE Prenatal androgen exposure could be a source of early programming, leading to the development of cardiometabolic diseases in later life. In this study, we aimed to examine cardiometabolic disturbances in males exposed to maternal androgen excess during their prenatal life. METHODS In this prospective population-based study, 409 male offspring with maternal hyperandrogenism (MHA), and 954 male offspring without MHA, as controls, were included. Both groups of male offspring were followed from the baseline to the date of the incidence of events, censoring, or end of the study period, whichever came first. Age-scaled unadjusted and adjusted Cox regression models were applied to assess the hazard ratios (HR) and 95% confidence intervals (CIs) for the association between MHA with pre-diabetes mellitus (Pre-DM), type 2 diabetes mellitus (T2DM), pre-hypertension (Pre-HTN), hypertension (HTN), dyslipidemia, overweight, and obesity in the offspring of both groups. Statistical analysis was performed using the STATA software package; the significance level was set at P < 0.05. RESULTS A higher risk of Pre-DM (adjusted HR: 1.46 (1.20, 1.78)) was observed in male offspring with MHA after adjustment for potential confounders, including body mass index, education, and physical activity. However, no significant differences were observed in the risk of T2DM, Pre-HTN, HTN, dyslipidemia, overweight, and obesity in males with MHA compared to controls in both the unadjusted and adjusted models. CONCLUSION Maternal androgen excess increases the risk of Pre-DM in male offspring in later life. More longitudinal studies with long enough follow-up are needed to clarify the effects of MHA on the cardiometabolic risk factors of male offspring in later life.
Collapse
Affiliation(s)
- M Farhadi-Azar
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 23 Arabi, Yaman Street, VelenjakTehran, 1985717413, Iran
| | - M Noroozzadeh
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 23 Arabi, Yaman Street, VelenjakTehran, 1985717413, Iran.
| | - M Ghahremani
- Department of Obstetrics and Gynecology, Education Program in Reproduction and Development, Monash University, Melbourne, VIC, Australia
| | - M Rahmati
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 23 Arabi, Yaman Street, VelenjakTehran, 1985717413, Iran
| | - M Saei Ghare Naz
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 23 Arabi, Yaman Street, VelenjakTehran, 1985717413, Iran
| | - F Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - F Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 23 Arabi, Yaman Street, VelenjakTehran, 1985717413, Iran
| |
Collapse
|
16
|
Lu S, Li Y, Qian Z, Zhao T, Feng Z, Weng X, Yu L. Role of the inflammasome in insulin resistance and type 2 diabetes mellitus. Front Immunol 2023; 14:1052756. [PMID: 36993972 PMCID: PMC10040598 DOI: 10.3389/fimmu.2023.1052756] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
The inflammasome is a protein complex composed of a variety of proteins in cells and which participates in the innate immune response of the body. It can be activated by upstream signal regulation and plays an important role in pyroptosis, apoptosis, inflammation, tumor regulation, etc. In recent years, the number of metabolic syndrome patients with insulin resistance (IR) has increased year by year, and the inflammasome is closely related to the occurrence and development of metabolic diseases. The inflammasome can directly or indirectly affect conduction of the insulin signaling pathway, involvement the occurrence of IR and type 2 diabetes mellitus (T2DM). Moreover, various therapeutic agents also work through the inflammasome to treat with diabetes. This review focuses on the role of inflammasome on IR and T2DM, pointing out the association and utility value. Briefly, we have discussed the main inflammasomes, including NLRP1, NLRP3, NLRC4, NLRP6 and AIM2, as well as their structure, activation and regulation in IR were described in detail. Finally, we discussed the current therapeutic options-associated with inflammasome for the treatment of T2DM. Specially, the NLRP3-related therapeutic agents and options are widely developed. In summary, this article reviews the role of and research progress on the inflammasome in IR and T2DM.
Collapse
Affiliation(s)
- Shen Lu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yanrong Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhaojun Qian
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Tiesuo Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaogang Weng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- *Correspondence: Lili Yu, ; Xiaogang Weng,
| | - Lili Yu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
- *Correspondence: Lili Yu, ; Xiaogang Weng,
| |
Collapse
|
17
|
Meng Q, Xu Y, Li Y, Wang Y. Novel studies on Drosophila melanogaster model reveal the roles of JNK-Jak/STAT axis and intestinal microbiota in insulin resistance. J Drug Target 2023; 31:261-268. [PMID: 36343203 DOI: 10.1080/1061186x.2022.2144869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The JNK pathway play a critical role in insulin resistance induced by a long-term high-sugar diet. However, the roles of up- and downstream molecules of the JNK pathway in insulin resistance are less known in vertebrates and invertebrates. As a classical organism in biological research, Drosophila melanogaster (D. melanogaster) has been widely applied to the studies of mechanism of insulin resistance. Based on previous studies, we found a novel predictive mechanism of the formation of insulin resistance in D. melanogaster. We found that JNK activated by high-sugar diet and dysregulated intestinal microbiota could mediate inflammation, and then the activated JNK released Upd3, which in turn stimulated Jak/STAT pathway to release ImpL2. ImpL2 can compete with Drosophila insulin-like peptides (Dilps) for binding with the insulin receptor and inhibit the activation of insulin pathway. In this study, we reviewed novel studies on the insulin signalling pathway based on the D. melanogaster model. The findings support our hypothesis. We, therefore, described how a long-term high-sugar diet disrupts intestinal microbiota to induce inflammation and the disruption of JNK-Jak/STAT axis. This description may offer some new clues to the formation of insulin resistance.
Collapse
Affiliation(s)
- Qinghao Meng
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yidong Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
18
|
Xu M, Xue H, Kong L, Lin L, Zheng G. Smilax china L. Polyphenols Improves Insulin Resistance and Obesity in High-fat Diet-induced Mice Through IRS/AKT-AMPK and NF-κB Signaling Pathways. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2023:10.1007/s11130-023-01052-y. [PMID: 36826691 DOI: 10.1007/s11130-023-01052-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Smilax china L. is an important herb used in traditional Chinese medicine. In this study, the mechanism of Smilax china L. polyphenols (SCP) on insulin resistance and anti-obesity in mice induced by a high-fat diet (HFD) was investigated. Fifty female mice were randomly divided into five groups: control, HFD and low, medium, and high doses of SCP for 70 d. SCP significantly decreased intraperitoneal adipose tissue index, body weight gain, liver lipids, and serum inflammatory factor levels. Blood glucose and insulin concentrations, as well as insulin resistance index in SCP, were significantly lower than those in HFD. In addition, SCP markedly up-regulated the gene expression of glucose transporter 4 (GLUT4), insulin receptor substrate 1 (IRS1), insulin receptor substrate 2 (IRS2), serine-threonine kinase (AKT), Acyl-CoA oxidase (ACO), and protein kinase A (PKA), and down-regulated the expression of mammalian target of rapamycin complex 1 (mTORC1), sterol-responsive element-binding protein-1c (SREBP1c), fatty acid synthase (FAS), 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGCR), and forkhead box protein O1 (FOXO1). SCP significantly increased the protein expression of AKT, GLUT4, AMP-activated protein kinase (AMPK), phosphorylated-AMPK (p-AMPK), phosphorylated-AKT (p-AKT), and uncoupling protein 1 (UCP-1), and decreased the expression of SREBP1c, FAS, HMGCR, phosphorylation of IKBα (p-IKBα), and nuclear factor kappa B subunit p65 (P65) in the liver. Overall, SCP effectively reduced HFD-induced insulin resistance and obesity in mice, partly through NF-κB and IRS/AKT-AMPK signaling pathways to regulate inflammatory factors. Therefore, SCP may improve lifestyle diseases.
Collapse
Affiliation(s)
- Meng Xu
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China
| | - Hui Xue
- Engineering Research Center of Biomass Conversion, Ministry of Education, Nanchang University, 330047, Nanchang, China
| | - Li Kong
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China
| | - Lezhen Lin
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China
| | - Guodong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China.
| |
Collapse
|
19
|
Wang JJ, Wang X, Li Q, Huang H, Zheng QL, Yao Q, Zhang J. Feto-placental endothelial dysfunction in Gestational Diabetes Mellitus under dietary or insulin therapy. BMC Endocr Disord 2023; 23:48. [PMID: 36814227 PMCID: PMC9948408 DOI: 10.1186/s12902-023-01305-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
OBJECTIVE Gestational diabetes mellitus (GDM) is a serious complication in pregnancy. Despite controlling the plasma glucose levels with dietary intervention (GDM-D) or insulin therapy (GDM-I), children born of diabetic mothers suffer more long-term complications from childhood to early adulthood. Placental circulation and nutrient exchange play a vital role in fetal development. Additionally, placental endothelial function is an indicator of vascular health, and plays an important role in maintaining placental circulation for nutrient exchange. This study was conducted to assess changes in fetal endothelial dysfunction in GDM under different interventions during pregnancy. METHODS The primary human umbilical vein endothelial cells (HUVECs) were obtained from normal pregnant women (n = 11), GDM-D (n = 14), and GDM-I (n = 12) patients. LC-MS/MS was used to identify differentially expressed proteins in primary HUVECs among the three groups, after which Bioinformatics analysis was performed. Glucose uptake, ATP level, apoptosis, and differentially expressed proteins were assessed to investigate changes in energy metabolism. RESULTS A total of 8174 quantifiable proteins were detected, and 142 differentially expressed proteins were identified after comparing patients with GDM-D/GDM-I and healthy controls. Of the 142, 64 proteins were upregulated while 77 were downregulated. Bioinformatics analysis revealed that the differentially expressed proteins were involved in multiple biological processes and signaling pathways related to cellular processes, biological regulation, and metabolic processes. According to the results from KEGG analysis, there were changes in the PI3K/AKT signaling pathway after comparing the three groups. In addition, there was a decrease in glucose uptake in the GDM-I (P < 0.01) group. In GDM-I, there was a significant decrease in the levels of glucose transporter 1 (GLUT1) and glucose transporter 3 (GLUT3). Moreover, glucose uptake was significantly decreased in GDM-I, although in GDM-D, there was only a decrease in the levels of GLUT1. ATP levels decreased in GDM-I (P < 0.05) and apoptosis occurred in both the GDM-D and GDM-I groups. Compared to the normal controls, the levels of phosphate AKT and phosphate AMPK over total AKT and AMPK were reduced in the GDM-I group. CONCLUSION In summary, endothelial dysfunction occurred in pregnancies with GDM even though the plasma glucose levels were controlled, and this dysfunction might be related to the degree of glucose tolerance. The energy dysfunction might be related to the regulation of the AKT/AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jing-Jing Wang
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China
| | - Xi Wang
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China
| | - Qian Li
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China
| | - Hua Huang
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China
| | - Qiao-Ling Zheng
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China
| | - Qin Yao
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China
| | - Jun Zhang
- Department of Clinical pharmacy, First Affiliated hospital of Kunming Medical University, Yunnan, China.
| |
Collapse
|
20
|
Nie G, Wan JJ, Jiang L, Hou SK, Peng W. Correlation Analysis between Uric Acid and Metabolic Syndrome in the Chinese Elderly Population: A Cross-Sectional Study. Int J Endocrinol 2023; 2023:8080578. [PMID: 36704419 PMCID: PMC9873429 DOI: 10.1155/2023/8080578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023] Open
Abstract
Background Currently, both metabolic syndrome and hyperuricaemia have attracted extensive attention in public health. The correlation between uric acid and metabolic syndrome is controversial. Research on the relationship between uric acid and metabolic syndrome in community-dwelling elderly people is relatively lacking. The purpose of this study is to explore the relationship between uric acid and metabolic syndrome in the community-dwelling elderly people. Design Cross-sectional study. Methods We collected the physical examination data of 1,267 elderly people in Gutian community in Wuhan and used SPSS IBM 25.0 for data analysis. Correlation and logistic regression analyses were performed, and ROC curves were drawn. Results The uric acid level of the nonmetabolic syndrome group was lower than that of the metabolic syndrome group (337.31 vs. 381.91 µmol/L; P < 0.05). Uric acid was positively correlated with systolic blood pressure (r = 0.177, P < 0.001), diastolic blood pressure (r = 0.135, P < 0.001), body mass index (r = 0.234, P < 0.001), waist circumference (r = 0.283, P < 0.001), and triglycerides (r = 0.217, P < 0.05). High-density lipoprotein cholesterol (r = -0.268, P < 0.001) showed the opposite trend. Logistic regression analysis results suggested that uric acid is a risk factor for metabolic syndrome. The result is described as exp (B) and 95% CI (1.003 [1.001, 1.005]). Based on the receiver operating characteristic curve, we found that the area under the curve of uric acid to diagnose metabolic syndrome was 0.64 (sensitivity: 79.3%, specificity: 45.1%). Conclusion We observed an association between uric acid levels and metabolic syndrome in the elderly Chinese population. The best threshold value for uric acid in predicting metabolic syndrome diagnosis was 314.5 μmol/l.
Collapse
Affiliation(s)
- Guqiao Nie
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing jing Wan
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Jiang
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu kai Hou
- Community Health Service Center, Gutian Street, Qiaokou District, Wuhan, Hubei, China
| | - Wen Peng
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Yang Z, Huang X, Zhang J, You K, Xiong Y, Fang J, Getachew A, Cheng Z, Yu X, Wang Y, Wu F, Wang N, Feng S, Lin X, Yang F, Chen Y, Wei H, Li YX. Hepatic DKK1-driven steatosis is CD36 dependent. Life Sci Alliance 2023; 6:e202201665. [PMID: 36410795 PMCID: PMC9679335 DOI: 10.26508/lsa.202201665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is prevalent worldwide; about 25% of NAFLD silently progress into steatohepatitis, in which some of them may develop into fibrosis, cirrhosis and liver failure. However, few drugs are available for NAFLD, partly because of an incomplete understanding of its pathogenic mechanisms. Here, using in vivo and in vitro gain- and loss-of-function approaches, we identified up-regulated DKK1 plays a pivotal role in high-fat diet-induced NAFLD and its progression. Mechanistic analysis reveals that DKK1 enhances the capacity of hepatocytes to uptake fatty acids through the ERK-PPARγ-CD36 axis. Moreover, DKK1 increased insulin resistance by activating the JNK signaling, which in turn exacerbates disorders of hepatic lipid metabolism. Our finding suggests that DKK1 may be a potential therapeutic and diagnosis candidate for NAFLD and metabolic disorder progression.
Collapse
Affiliation(s)
- Zhen Yang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinping Huang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaye Zhang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kai You
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Xiong
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ji Fang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Anteneh Getachew
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ziqi Cheng
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaorui Yu
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yan Wang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Feima Wu
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ning Wang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shufen Feng
- Department of Gastroenterology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xianhua Lin
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fan Yang
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yan Chen
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hongcheng Wei
- Department of Gastroenterology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yin-Xiong Li
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| |
Collapse
|
22
|
Guo Z, Yamaguchi R. Machine learning methods for protein-protein binding affinity prediction in protein design. FRONTIERS IN BIOINFORMATICS 2022; 2:1065703. [PMID: 36591334 PMCID: PMC9800603 DOI: 10.3389/fbinf.2022.1065703] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Protein-protein interactions govern a wide range of biological activity. A proper estimation of the protein-protein binding affinity is vital to design proteins with high specificity and binding affinity toward a target protein, which has a variety of applications including antibody design in immunotherapy, enzyme engineering for reaction optimization, and construction of biosensors. However, experimental and theoretical modelling methods are time-consuming, hinder the exploration of the entire protein space, and deter the identification of optimal proteins that meet the requirements of practical applications. In recent years, the rapid development in machine learning methods for protein-protein binding affinity prediction has revealed the potential of a paradigm shift in protein design. Here, we review the prediction methods and associated datasets and discuss the requirements and construction methods of binding affinity prediction models for protein design.
Collapse
Affiliation(s)
- Zhongliang Guo
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan,Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan,*Correspondence: Rui Yamaguchi,
| |
Collapse
|
23
|
Abdollahi M, Kato M, Lanting L, Tunduguru R, Wang M, Wang Y, Fueger PT, Wang Q, Huang W, Natarajan R. miR-379 mediates insulin resistance and obesity through impaired angiogenesis and adipogenesis regulated by ER stress. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:115-130. [PMID: 36250205 PMCID: PMC9535382 DOI: 10.1016/j.omtn.2022.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/15/2022] [Indexed: 01/29/2023]
Abstract
We investigated the role of microRNA (miR-379) in the pathogenesis of obesity, adipose tissue dysfunction, and insulin resistance (IR). We used miR-379 knockout (miR-379KO) mice to test whether loss of miR-379 affects high-fat diet (HFD)-induced obesity and IR via dysregulation of key miR-379 targets in adipose tissue. Increases in body weight, hyperinsulinemia, and IR in wild-type (WT)-HFD mice were significantly attenuated in miR-379KO-HFD mice with some sex differences. Relative to control chow-fed mice, in WT-HFD mice, expression of miR-379 and C/EBP homologous protein (Chop) (pro-endoplasmic reticulum [ER] stress) and inflammation in perigonadal white adipose tissue (gWAT) were increased, whereas adipogenic genes and miR-379 target genes (Vegfb and Edem3) were decreased. These changes, as well as key parameters of brown adipose tissue dysfunction (including mitochondrial defects), were significantly attenuated in miR-379KO-HFD mice. WAT from obese human subjects with and without type 2 diabetes showed increased miR-379 and decreased miR-379 target genes. In cultured 3T3L1 pre-adipocytes, miR-379 inhibitors increased miR-379 targets and adipogenic genes. These data suggest that miR-379 plays an important role in HFD-induced obesity through increased adipose inflammation, mitochondrial dysfunction, and ER stress as well as impaired adipogenesis and angiogenesis. miR-379 inhibitors may be developed as novel therapies for obesity and associated complications.
Collapse
Affiliation(s)
- Maryam Abdollahi
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mitsuo Kato
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Linda Lanting
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Ragadeepthi Tunduguru
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mei Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Yangmeng Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Patrick T. Fueger
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Qiong Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
24
|
Acosta-Martinez M, Cabail MZ. The PI3K/Akt Pathway in Meta-Inflammation. Int J Mol Sci 2022; 23:ijms232315330. [PMID: 36499659 PMCID: PMC9740745 DOI: 10.3390/ijms232315330] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Obesity is a global epidemic representing a serious public health burden as it is a major risk factor for the development of cardiovascular disease, stroke and all-cause mortality. Chronic low-grade systemic inflammation, also known as meta-inflammation, is thought to underly obesity's negative health consequences, which include insulin resistance and the development of type 2 diabetes. Meta-inflammation is characterized by the accumulation of immune cells in adipose tissue, a deregulation in the synthesis and release of adipokines and a pronounced increase in the production of proinflammatory factors. In this state, the infiltration of macrophages and their metabolic activation contributes to complex paracrine and autocrine signaling, which sustains a proinflammatory microenvironment. A key signaling pathway mediating the response of macrophages and adipocytes to a microenvironment of excessive nutrients is the phosphoinositide 3-kinase (PI3K)/Akt pathway. This multifaceted network not only transduces metabolic information but also regulates macrophages' intracellular changes, which are responsible for their phenotypic switch towards a more proinflammatory state. In the present review, we discuss how the crosstalk between macrophages and adipocytes contributes to meta-inflammation and provide an overview on the involvement of the PI3K/Akt signaling pathway, and how its impairment contributes to the development of insulin resistance.
Collapse
Affiliation(s)
- Maricedes Acosta-Martinez
- Department of Physiology and Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maria Zulema Cabail
- Biological Science Department, State University of New York-College at Old Westbury, Old Westbury, NY 11568, USA
- Correspondence:
| |
Collapse
|
25
|
Gáliková M, Klepsatel P. Ion transport peptide regulates energy intake, expenditure, and metabolic homeostasis in Drosophila. Genetics 2022; 222:iyac150. [PMID: 36190340 PMCID: PMC9713441 DOI: 10.1093/genetics/iyac150] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/21/2022] [Indexed: 12/13/2022] Open
Abstract
In mammals, energy homeostasis is regulated by the antagonistic action of hormones insulin and glucagon. However, in contrast to the highly conserved insulin, glucagon is absent in most invertebrates. Although there are several endocrine regulators of energy expenditure and catabolism (such as the adipokinetic hormone), no single invertebrate hormone with all of the functions of glucagon has been described so far. Here, we used genetic gain- and loss-of-function experiments to show that the Drosophila gene Ion transport peptide (ITP) codes for a novel catabolic regulator that increases energy expenditure, lowers fat and glycogen reserves, and increases glucose and trehalose. Intriguingly, Ion transport peptide has additional functions reminiscent of glucagon, such as inhibition of feeding and transit of the meal throughout the digestive tract. Furthermore, Ion transport peptide interacts with the well-known signaling via the Adipokinetic hormone; Ion transport peptide promotes the pathway by stimulating Adipokinetic hormone secretion and transcription of the receptor AkhR. The genetic manipulations of Ion transport peptide on standard and Adipokinetic hormone-deficient backgrounds showed that the Adipokinetic hormone peptide mediates the hyperglycemic and hypertrehalosemic effects of Ion transport peptide, while the other metabolic functions of Ion transport peptide seem to be Adipokinetic hormone independent. In addition, Ion transport peptide is necessary for critical processes such as development, starvation-induced foraging, reproduction, and average lifespan. Altogether, our work describes a novel master regulator of fly physiology with functions closely resembling mammalian glucagon.
Collapse
Affiliation(s)
- Martina Gáliková
- Institute of Zoology, Slovak Academy of Sciences, 845 06 Bratislava, Slovakia
- Department of Zoology, Stockholm University, 106 91 Stockholm, Sweden
| | - Peter Klepsatel
- Institute of Zoology, Slovak Academy of Sciences, 845 06 Bratislava, Slovakia
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| |
Collapse
|
26
|
Szukiewicz D, Trojanowski S, Kociszewska A, Szewczyk G. Modulation of the Inflammatory Response in Polycystic Ovary Syndrome (PCOS)-Searching for Epigenetic Factors. Int J Mol Sci 2022; 23:ijms232314663. [PMID: 36498989 PMCID: PMC9736994 DOI: 10.3390/ijms232314663] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Despite its incidence, the syndrome is poorly understood and remains underdiagnosed, and female patients are diagnosed with a delay. The heterogenous nature of this complex disorder results from the combined occurrence of genetic, environmental, endocrine, and behavioral factors. Primary clinical manifestations of PCOS are derived from the excess of androgens (anovulation, polycystic ovary morphology, lack of or scanty, irregular menstrual periods, acne and hirsutism), whereas the secondary manifestations include multiple metabolic, cardiovascular, and psychological disorders. Dietary and lifestyle factors play important roles in the development and course of PCOS, which suggests strong epigenetic and environmental influences. Many studies have shown a strong association between PCOS and chronic, low-grade inflammation both in the ovarian tissue and throughout the body. In the vast majority of PCOS patients, elevated values of inflammatory markers or their gene markers have been reported. Development of the vicious cycle of the chronic inflammatory state in PCOS is additionally stimulated by hyperinsulinemia and obesity. Changes in DNA methylation, histone acetylation and noncoding RNA levels are presented in this review in the context of oxidative stress, reactive oxygen species, and inflammatory signaling in PCOS. Epigenetic modulation of androgenic activity in response to inflammatory signaling is also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
- Correspondence:
| | - Seweryn Trojanowski
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Anna Kociszewska
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
27
|
Li X, Geng-Ji JJ, Quan YY, Qi LM, Sun Q, Huang Q, Jiang HM, Sun ZJ, Liu HM, Xie X. Role of potential bioactive metabolites from traditional Chinese medicine for type 2 diabetes mellitus: An overview. Front Pharmacol 2022; 13:1023713. [PMID: 36479195 PMCID: PMC9719995 DOI: 10.3389/fphar.2022.1023713] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/07/2022] [Indexed: 11/14/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease with persistent hyperglycemia primarily caused by insulin resistance (IR). The number of diabetic patients globally has been rising over the past decades. Although significant progress has been made in treating diabetes mellitus (DM), existing clinical drugs for diabetes can no longer fully meet patients when they face complex and huge clinical treatment needs. As a traditional and effective medical system, traditional Chinese medicine (TCM) has a unique understanding of diabetes treatment and has developed many classic and practical prescriptions targeting DM. With modern medicine and pharmacy advancements, researchers have discovered that various bioactive metabolites isolated from TCM show therapeutic on DM. Compared with existing clinical drugs, these bioactive metabolites demonstrate promising prospects for treating DM due to their excellent biocompatibility and fewer adverse reactions. Accordingly, these valuable metabolites have attracted the interest of researchers worldwide. Despite the abundance of research works and specialized-topic reviews published over the past years, there is a lack of updated and systematic reviews concerning this fast-growing field. Therefore, in this review, we summarized the bioactive metabolites derived from TCM with the potential treatment of T2DM by searching several authoritative databases such as PubMed, Web of Science, Wiley Online Library, and Springer Link. For the convenience of readers, the content is divided into four parts according to the structural characteristics of these valuable compounds (flavonoids, terpenoids, alkaloids, and others). Meanwhile, the detailed mechanism and future directions of these promising compounds curing DM are also summarized in the related sections. We hope this review inspires increasingly valuable and significant research focusing on potential bioactive metabolites from TCM to treat DM in the future.
Collapse
Affiliation(s)
- Xiang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, School of Pharmacy and College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia-Jia Geng-Ji
- State Key Laboratory of Southwestern Chinese Medicine Resources, State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, School of Pharmacy and College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun-Yun Quan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Lu-Ming Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, School of Pharmacy and College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Sun
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qun Huang
- Department of Ophthalmology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai-Mei Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, School of Pharmacy and College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zi-Jian Sun
- Sichuan Ant Recommendation Biotechnology Co., Ltd., Chengdu, Sichuan, China
| | - Hong-Mei Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xin Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, School of Pharmacy and College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
28
|
Ahmed ES, Mohamed HE, Farrag MA. Luteolin loaded on zinc oxide nanoparticles ameliorates non-alcoholic fatty liver disease associated with insulin resistance in diabetic rats via regulation of PI3K/AKT/FoxO1 pathway. Int J Immunopathol Pharmacol 2022; 36:3946320221137435. [PMID: 36319192 PMCID: PMC9630902 DOI: 10.1177/03946320221137435] [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: 11/07/2022] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is a worldwide health problem with high prevalence and morbidity associated with obesity, insulin resistance, type 2 diabetes mellitus (T2DM), and dyslipidemia. Nano-formulation of luteolin with Zn oxide in the form of Lut/ZnO NPs may improve the anti-diabetic property of each alone and ameliorate the insulin resistance thus management of NAFLD. This study aimed to measure the efficiency of Lut/ZnO NPs against insulin resistance coupled with NAFLD and T2DM. METHODS A diabetic rat model with NAFLD was induced by a high-fat diet and streptozotocin (30 mg/kg I.P). Serum diabetogenic markers levels, lipid profile, and activity of liver enzymes were measured beside liver oxidative stress markers. Moreover, the hepatic expressions of PI3K/AKT/FoxO1/SERBP1c as well as heme oxygenase-1 were measured beside the histopathological examination. RESULTS Lut/ZnO NPs treatment effectively reduced hyperglycemia, hyperinsulinemia, and ameliorated insulin resistance. Additionally, Lut/ZnO NPs improved the hepatic functions, the antioxidant system, and reduced the oxidative stress markers. Furthermore, the lipid load in the liver, as well as the circulating TG and TC, was minified via the suppression of lipogenesis and gluconeogenesis. Moreover, Lut/ZnO NPs activated the PI3K/AKT signaling pathway, hence inactivating FoxO1, therefore enhancing the hepatic cells' insulin sensitivity. CONCLUSION Lut/ZnO NPs have a hepatoprotective effect and may relieve the progression of NAFLD by alleviating insulin resistance, ameliorating the antioxidant status, and regulating the insulin signal pathway.
Collapse
Affiliation(s)
- Esraa Sa Ahmed
- Radiation Biology Research, National Center for Radiation Research and Technology, 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hebatallah E Mohamed
- Radiation Biology Research, National Center for Radiation Research and Technology, 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mostafa A Farrag
- Radiation Biology Research, National Center for Radiation Research and Technology, 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
29
|
Miura I, Okada K, Ishii A, Warabi E, Watahiki T, To K, Shimano H, Ariizumi S, Shoda J. p62/Sqstm1 rescue in muscle retards the progression of steatohepatitis in p62/Sqstm1-null mice fed a high-fat diet. Front Physiol 2022; 13:993995. [PMID: 36439272 PMCID: PMC9692207 DOI: 10.3389/fphys.2022.993995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction: Obesity is a risk factor for many diseases because it leads to a reduction in skeletal muscle mass and promotes insulin resistance. p62/Sqstm1-knockout mice are a model of metabolic syndrome; show obesity, insulin resistance, and non-alcoholic fatty liver (NAFL); and develop non-alcoholic steatohepatitis (NASH) in response to the feeding of a high-fat diet (HFD). These phenotypes suggest that muscle p62 may prevent obesity-induced muscle dysfunction. In the present study, we aimed to determine the effects of muscle p62 on skeletal muscle mass, muscle strength, insulin resistance, and NASH pathology. Methods: We generated muscle-specific p62 gene rescue mice (p62-mRes), which express p62 only in muscle and were derived from p62-knock out mice (p62KIKI) using the cre/loxp system. p62KIKI and p62-mRes mice were fed an HFD for 20 weeks and their phenotypes were compared. Results: HFD-feeding caused severe obesity in both p62KIKI and p62-mRes mice, but there was no effect of muscle p62 on body mass. Limb skeletal muscle mass, grip strength, and the cross-sectional area of muscle fibers were higher in p62-mRes mice than in p62KIKI. The glucose tolerance and insulin sensitivity of the p62-mRes mice were also superior. The protein expression of mechanistic target of rapamycin, which promotes muscle protein synthesis, and GLUT4, a glucose transporter in skeletal muscle, were higher in the p62-mRes mice. p62KIKI mice developed severe NASH when fed an HFD, but the progression of NASH was retarded by p62 gene rescue in muscle, and the expression of Tgf-β1, which encodes a factor that promotes hepatic fibrosis, was reduced. Conclusion: Rescue of muscle-specific p62 in the whole-body p62 knock-out mice ameliorates the insulin resistance and retards the progression of NASH caused by systemic p62 ablation.
Collapse
Affiliation(s)
- Ikuru Miura
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Kosuke Okada
- Department of Medical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akiko Ishii
- Department of Internal Medicine (Neurology), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Eiji Warabi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takahisa Watahiki
- Doctoral Program in Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Keii To
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shunichi Ariizumi
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Shinjuku, Japan
| | - Junichi Shoda
- Department of Medical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
30
|
Matrone A, Basolo A, Santini F, Elisei R. Understanding the effect of obesity on papillary thyroid cancer: is there a need for tailored diagnostic and therapeutic management? Expert Rev Endocrinol Metab 2022; 17:475-484. [PMID: 36203362 DOI: 10.1080/17446651.2022.2131529] [Citation(s) in RCA: 2] [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: 02/24/2022] [Accepted: 09/29/2022] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Several studies have focused on the relationship between obesity and differentiated thyroid carcinoma (DTC), particularly papillary histotype (PTC). However, the association of obesity with both incidence and aggressiveness of PTC is still incompletely understood. AREAS COVERED We reviewed the mechanisms underlying the cross talk between obesity and thyroid carcinomas and described the most recent evidence evaluating the effect of obesity on the development of PTC, as well as the impact of excessive body weight on the clinicopathologic features and outcome of this type of cancer. EXPERT OPINION Available evidence suggests that excessive body weight is linked with a higher risk of getting PTC, while its impact on the aggressiveness of the disease, if present, is still not clear. Therefore, while attention should be paid to discover thyroid cancer in patients with obesity earlier, once diagnosed it should be managed following a conventional workup as in normal weight patients, based on the clinical presentation of the disease and including active surveillance if appropriate, as recommended by referral guidelines.
Collapse
Affiliation(s)
- Antonio Matrone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa - via Paradisa 2, Pisa, Italy
| | - Alessio Basolo
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa - via Paradisa 2, Pisa, Italy
| | - Ferruccio Santini
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa - via Paradisa 2, Pisa, Italy
| | - Rossella Elisei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa - via Paradisa 2, Pisa, Italy
| |
Collapse
|
31
|
Liu Y, Feng J, Pan H, Zhang X, Zhang Y. Genetically engineered bacterium: Principles, practices, and prospects. Front Microbiol 2022; 13:997587. [PMID: 36312915 PMCID: PMC9606703 DOI: 10.3389/fmicb.2022.997587] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022] Open
Abstract
Advances in synthetic biology and the clinical application of bacteriotherapy enable the use of genetically engineered bacteria (GEB) to combat various diseases. GEB act as a small 'machine factory' in the intestine or other tissues to continuously produce heterologous proteins or molecular compounds and, thus, diagnose or cure disease or work as an adjuvant reagent for disease treatment by regulating the immune system. Although the achievements of GEBs in the treatment or adjuvant therapy of diseases are promising, the practical implementation of this new therapeutic modality remains a grand challenge, especially at the initial stage. In this review, we introduce the development of GEBs and their advantages in disease management, summarize the latest research advances in microbial genetic techniques, and discuss their administration routes, performance indicators and the limitations of GEBs used as platforms for disease management. We also present several examples of GEB applications in the treatment of cancers and metabolic diseases and further highlight their great potential for clinical application in the near future.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Hangcheng Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuwei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Yunlei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Kábelová A, Malínská H, Marková I, Hűttl M, Chylíková B, Šeda O. Quercetin supplementation alters adipose tissue and hepatic transcriptomes and ameliorates adiposity, dyslipidemia, and glucose intolerance in adult male rats. Front Nutr 2022; 9:952065. [PMID: 36245490 PMCID: PMC9558266 DOI: 10.3389/fnut.2022.952065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Quercetin, a flavonoid present in many fruits and vegetables, exhibits beneficial effects toward abnormalities related to metabolic syndrome. In this study, to further investigate metabolic and transcriptomic responses to quercetin supplementation, we used a genetic model of metabolic syndrome. Adult male rats of the PD/Cub strain were fed either a high-sucrose diet (HSD; control PD rats) or HSD fortified with quercetin (10 g quercetin/kg diet; PD-Q rats). Morphometric and metabolic parameters, along with transcriptomic profiles of the liver and retroperitoneal fat, were assessed. The relative weights of epididymal and retroperitoneal fat were significantly decreased in quercetin-treated animals. Furthermore, a smaller area under the glycemic curve along with a decreased level of fasting insulin were detected in PD-Q rats. While no changes in total cholesterol levels were observed, the overall level of triglycerides decreased in the serum and the liver of the PD-Q rats. The transcriptomic profile of the liver and the adipose tissue corroborated the metabolic and morphometric findings, revealing the pattern consistent with insulin-sensitizing changes, with major regulator nodes being Pparg, Adipoq, Nos2, and Mir378. In conclusion, quercetin supplementation improves abnormalities related to metabolic syndrome, namely adiposity, dyslipidemia and glucose intolerance.
Collapse
Affiliation(s)
- Adéla Kábelová
- First Faculty of Medicine, Institute of Biology and Medical Genetics, General University Hospital, Charles University, Prague, Czechia
| | - Hana Malínská
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Irena Marková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Martina Hűttl
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Blanka Chylíková
- First Faculty of Medicine, Institute of Biology and Medical Genetics, General University Hospital, Charles University, Prague, Czechia
| | - Ondřej Šeda
- First Faculty of Medicine, Institute of Biology and Medical Genetics, General University Hospital, Charles University, Prague, Czechia
- *Correspondence: Ondřej Šeda,
| |
Collapse
|
33
|
Gorgisen G, Aydin M, Mboma O, Gökyildirim MY, Chao CM. The Role of Insulin Receptor Substrate Proteins in Bronchopulmonary Dysplasia and Asthma: New Potential Perspectives. Int J Mol Sci 2022; 23:ijms231710113. [PMID: 36077511 PMCID: PMC9456457 DOI: 10.3390/ijms231710113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 01/12/2023] Open
Abstract
Insulin receptor substrates (IRSs) are proteins that are involved in signaling through the insulin receptor (IR) and insulin-like growth factor (IGFR). They can also interact with other receptors including growth factor receptors. Thus, they represent a critical node for the transduction and regulation of multiple signaling pathways in response to extracellular stimuli. In addition, IRSs play a central role in processes such as inflammation, growth, metabolism, and proliferation. Previous studies have highlighted the role of IRS proteins in lung diseases, in particular asthma. Further, the members of the IRS family are the common proteins of the insulin growth factor signaling cascade involved in lung development and disrupted in bronchopulmonary dysplasia (BPD). However, there is no study focusing on the relationship between IRS proteins and BPD yet. Unfortunately, there is still a significant gap in knowledge in this field. Thus, in this review, we aimed to summarize the current knowledge with the major goal of exploring the possible roles of IRS in BPD and asthma to foster new perspectives for further investigations.
Collapse
Affiliation(s)
- Gokhan Gorgisen
- Department of Medical Genetics, Faculty of Medicine, Van Yüzüncü Yil University, Van 65080, Turkey
| | - Malik Aydin
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
- Center for Child and Adolescent Medicine, Center for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| | - Olivier Mboma
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
- Center for Child and Adolescent Medicine, Center for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| | - Mira Y. Gökyildirim
- Department of Pediatrics, University Medical Center Rostock, University of Rostock, 18057 Rostock, Germany
| | - Cho-Ming Chao
- Department of Pediatrics, University Medical Center Rostock, University of Rostock, 18057 Rostock, Germany
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, 35390 Giessen, Germany
- Correspondence: ; Tel.: +49-641-9946735
| |
Collapse
|
34
|
Akter S, Akhter H, Chaudhury HS, Rahman MH, Gorski A, Hasan MN, Shin Y, Rahman MA, Nguyen MN, Choi TG, Kim SS. Dietary carbohydrates: Pathogenesis and potential therapeutic targets to obesity-associated metabolic syndrome. Biofactors 2022; 48:1036-1059. [PMID: 36102254 DOI: 10.1002/biof.1886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023]
Abstract
Metabolic syndrome (MetS) is a common feature in obesity, comprising a cluster of abnormalities including abdominal fat accumulation, hyperglycemia, hyperinsulinemia, dyslipidemia, and hypertension, leading to diabetes and cardiovascular diseases (CVD). Intake of carbohydrates (CHO), particularly a sugary diet that rapidly increases blood glucose, triglycerides, and blood pressure levels is the predominant determining factor of MetS. Complex CHO, on the other hand, are a stable source of energy taking a longer time to digest. In particular, resistant starch (RS) or soluble fiber is an excellent source of prebiotics, which alter the gut microbial composition, which in turn improves metabolic control. Altering maternal CHO intake during pregnancy may result in the child developing MetS. Furthermore, lifestyle factors such as physical inactivity in combination with dietary habits may synergistically influence gene expression by modulating genetic and epigenetic regulators transforming childhood obesity into adolescent metabolic disorders. This review summarizes the common pathophysiology of MetS in connection with the nature of CHO, intrauterine nutrition, genetic predisposition, lifestyle factors, and advanced treatment approaches; it also emphasizes how dietary CHO may act as a key element in the pathogenesis and future therapeutic targets of obesity and MetS.
Collapse
Affiliation(s)
- Salima Akter
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medical Biotechnology, Bangladesh University of Health Sciences, Dhaka 1216, Bangladesh
| | - Hajara Akhter
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| | - Habib Sadat Chaudhury
- Department of Biochemistry, International Medical College Hospital, Tongi 1711, Bangladesh
| | - Md Hasanur Rahman
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Andrew Gorski
- Department of Philosophy in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | | | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Md Ataur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Global Biotechnology & Biomedical Research Network (GBBRN), Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Minh Nam Nguyen
- Research Center for Genetics and Reproductive Health, School of Medicine, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung-Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Pristine Pharmaceuticals, Patuakhali 8600, Bangladesh
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
35
|
Alpha-Ketoglutarate Alleviates Neuronal Apoptosis Induced by Central Insulin Resistance through Inhibiting S6K1 Phosphorylation after Subarachnoid Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9148257. [PMID: 36062190 PMCID: PMC9436633 DOI: 10.1155/2022/9148257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/18/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022]
Abstract
Neuronal apoptosis after subarachnoid hemorrhage (SAH) is believed to play an important role in early brain injury after SAH. The energy metabolism of neuron is closely related to its survival. The transient hyperglycemia caused by insulin resistance (IR) after SAH seriously affects the prognosis of patients. However, the specific mechanisms of IR after SAH are still not clear. Studies have shown that α-KG takes part in the regulation of IR and cell apoptosis. In this study, we aim to investigate whether α-KG can reduce IR after SAH, improve the disorder of neuronal glucose metabolism, alleviate neuronal apoptosis, and ultimately play a neuroprotective role in SAH-induced EBI. We first measured α-KG levels in the cerebrospinal fluid (CSF) of patients with SAH. Then, we established a SAH model through hemoglobin (Hb) stimulation with HT22 cells for further mechanism research. Furthermore, an in vivo SAH model in mice was established by endovascular perforation. Our results showed that α-KG levels in CSF significantly increased in SAH patients and could be used as a potential prognostic biomarker. In in vitro model of SAH, we found that α-KG not only inhibited IR-induced reduction of glucose uptake in neurons after SAH but also alleviated SAH-induced neuronal apoptosis. Mechanistically, we found that α-KG inhibits neuronal IR by inhibiting S6K1 activation after SAH. Moreover, neuronal apoptosis significantly increased when glucose uptake was reduced. Furthermore, our results demonstrated that α-KG could also alleviate neuronal apoptosis in vivo SAH model. In conclusion, our study suggests that α-KG alleviates apoptosis by inhibiting IR induced by S6K1 activation after SAH.
Collapse
|
36
|
Xi R, Wan Y, Yang L, Zhang J, Yang L, Yang S, Chai R, Mu F, Sun Q, Yan R, Wu Z, Li S. Investigating Celastrol's Anti-DCM Targets and Mechanisms via Network Pharmacology and Experimental Validation. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7382130. [PMID: 35845929 PMCID: PMC9278495 DOI: 10.1155/2022/7382130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022]
Abstract
Methods Data from TCMSP and GEO databases were utilized to identify targets for Celastrol on DCM. The relationship between the major targets and conventional glycolipid metabolism was obtained with Spearman correlation analysis. Experiments on animals were conducted utilizing healthy control (HC), low-dose Celastrol interventions (CL), and no intervention groups (NC), all of which had 8 SD rats in each group. To study alterations in signaling molecules, RT-PCR was performed. Results There were 76 common targets and 5 major targets for Celastrol-DCM. Celastrol have been found to regulate AGE-RAGE, TNF, MAPK, TOLL-like receptors, insulin resistance, and other signaling pathways, and they are closely linked to adipocytokines, fatty acid metabolism, glycolipid biosynthesis, and glycosylphosphati-dylinositol biosynthesis on DCM. These five major targets have been found to regulate these pathways. Experiments on rats indicated that P38 MAPK was considerably elevated in the cardiac tissue from rats in the CL and NC groups compared to the HC group, and the difference was statistically significant (P < 0.01). Significant differences were seen between the CL and NC groups in P38 MAPK levels, with a statistical significance level of less than 0.05. Conclusion Celastrol may play a role in reversing energy remodeling, anti-inflammation, and oxidative stress via modulating p38 protein expression in the MAPK pathway, which have been shown in the treatment of DCM.
Collapse
Affiliation(s)
- Rui Xi
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongxin Wan
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lihong Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jingying Zhang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Liu Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shuai Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Chai
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fengchen Mu
- Department of Vascular Medicine, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, China
| | - Qiting Sun
- Department of Nuclear Medicine, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, China
| | - Rui Yan
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
37
|
Rastrelli G, Cipriani S, Lotti F, Cellai I, Comeglio P, Filippi S, Boddi V, Della Camera PA, Santi R, Boni L, Nesi G, Serni S, Gacci M, Maggi M, Vignozzi L. Testosterone does not affect lower urinary tract symptoms while improving markers of prostatitis in men with benign prostatic hyperplasia: a randomized clinical trial. J Endocrinol Invest 2022; 45:1413-1425. [PMID: 35298833 PMCID: PMC9184417 DOI: 10.1007/s40618-022-01776-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/18/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Benign Prostatic Hyperplasia (BPH) is a result of prostate inflammation, frequently occurring in metabolic syndrome (MetS). Low testosterone is common in MetS. A randomized clinical trial was designed to evaluate if 24 weeks of testosterone therapy (TTh) in BPH men with MetS and low testosterone improve urinary symptoms and prostate inflammation. METHODS One-hundred-twenty men with MetS waitlisted for BPH surgery were enrolled. They were categorized into normal testosterone (TT ≥ 12 nmol/L and cFT ≥ 225 pmol/L; n = 48) and testosterone deficient (TD) (TT < 12 nmol/L and/or cFT < 225 pmol/L; n = 72) then randomized to testosterone gel 2% (5 g/daily) or placebo for 24 weeks. At baseline and follow-up, questionnaires for urinary symptoms and trans-rectal ultrasound were performed. Prostate tissue was collected for molecular and histopathological analyses. RESULTS No differences in the improvement of urinary symptoms were found between TTh and placebo (OR [95% CI] 0.96 [0.39; 2.37]). In TD + TTh, increase in prostate but not adenoma volume was observed (2.64 mL [0.07; 5.20] and 1.82 mL [- 0.46; 0.41], respectively). Ultrasound markers of inflammation were improved. In a subset of 61 men, a hyper-expression of several pro-inflammatory genes was found in TD + placebo when compared with normal testosterone. TTh was able to counteract this effect. For 80 men, the inflammatory infiltrate was higher in TD + placebo than in normal testosterone (0.8 points [0.2; 1.4]) and TD + TTh men (0.9 points [0.2; 1.5]). CONCLUSIONS Twenty-four weeks of TTh in TD men with BPH and MetS improves ultrasound, molecular and histological proxies of prostate inflammation. This does not result in symptom improvement.
Collapse
Affiliation(s)
- G Rastrelli
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - S Cipriani
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - F Lotti
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - I Cellai
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - P Comeglio
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - S Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of Reproduction, Department of Neuroscience, Psychology, Pharmacology and Child Health, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - V Boddi
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - P A Della Camera
- Urology Unit, Azienda Ospedaliera Universitaria Careggi, Largo Piero Palagi, 1, 50139, Florence, Italy
| | - R Santi
- Pathological Anatomy Unit, Careggi University Hospital, Largo Piero Palagi, 1, 50139, Florence, Italy
| | - L Boni
- Unit of Clinical Epidemiology, IRCCS Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - G Nesi
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - S Serni
- Urology Unit, Azienda Ospedaliera Universitaria Careggi, Largo Piero Palagi, 1, 50139, Florence, Italy
| | - M Gacci
- Urology Unit, Azienda Ospedaliera Universitaria Careggi, Largo Piero Palagi, 1, 50139, Florence, Italy
| | - M Maggi
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
- Istituto Nazionale Biostrutture e Biosistemi, Viale delle Medaglie d'Oro, 305, 00136, Rome, Italy
| | - L Vignozzi
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy.
- Istituto Nazionale Biostrutture e Biosistemi, Viale delle Medaglie d'Oro, 305, 00136, Rome, Italy.
| |
Collapse
|
38
|
Nurcahyanti ADR, Cokro F, Wulanjati MP, Mahmoud MF, Wink M, Sobeh M. Curcuminoids for Metabolic Syndrome: Meta-Analysis Evidences Toward Personalized Prevention and Treatment Management. Front Nutr 2022; 9:891339. [PMID: 35757255 PMCID: PMC9218575 DOI: 10.3389/fnut.2022.891339] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
The metabolic syndrome (MS) is a multifactorial syndrome associated with a significant economic burden and healthcare costs. MS management often requires multiple treatments (polydrug) to ameliorate conditions such as diabetes mellitus, insulin resistance, obesity, cardiovascular diseases, hypertension, and non-alcoholic fatty liver disease (NAFLD). However, various therapeutics and possible drug-drug interactions may also increase the risk of MS by altering lipid and glucose metabolism and promoting weight gain. In addition, the medications cause side effects such as nausea, flatulence, bloating, insomnia, restlessness, asthenia, palpitations, cardiac arrhythmias, dizziness, and blurred vision. Therefore, is important to identify and develop new safe and effective agents based on a multi-target approach to treat and manage MS. Natural products, such as curcumin, have multi-modalities to simultaneously target several factors involved in the development of MS. This review discusses the recent preclinical and clinical findings, and up-to-date meta-analysis from Randomized Controlled Trials regarding the effects of curcumin on MS, as well as the metabonomics and a pharma-metabolomics outlook considering curcumin metabolites, the gut microbiome, and environment for a complementary personalized prevention and treatment for MS management.
Collapse
Affiliation(s)
- Agustina Dwi Retno Nurcahyanti
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Fonny Cokro
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Martha P Wulanjati
- Research Division for Natural Products Technology (BPTBA), National Research and Innovation Agency (BRIN), Yogyakarta, Indonesia
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg University, Heidelberg, Germany
| | - Mansour Sobeh
- AgroBioSciences Department, Mohammed VI Polytechnic University, Ben-Guerir, Morocco
| |
Collapse
|
39
|
Kinin B1 receptor deficiency protects mice fed by cafeteria diet from abnormal glucose homeostasis. PLoS One 2022; 17:e0267845. [PMID: 35617279 PMCID: PMC9135186 DOI: 10.1371/journal.pone.0267845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/14/2022] [Indexed: 11/24/2022] Open
Abstract
The kallikrein–kinin system has been implicated in body weight and glucose homeostasis. Their major effectors act by binding to the kinin B2 and B1 receptors. It was assessed the role of the kinin B1 receptor in weight and glucose homeostasis in B1 receptor knockout mice (B1RKO) subjected to a cafeteria diet (CAF). Wild-type (WT) and B1RKO male mice (C57BL/6 background; 8 weeks old) were fed a standard diet (SD) or CAF for 14 weeks, ad libitum, and four groups were formed: WT-SD; B1RKO-SD; WT-CAF; B1RKO-CAF. Body weight and food intake were assessed weekly. It was performed glucose tolerance (GTT) and insulin tolerance tests (ITT), and HOMA-IR, HOMA-β and HOMA-β* 1/HOMA-IR were calculated. Islets from WT and B1RKO were isolated in order to measure the insulin secretion. Western blot was used to assess the hepatic AKT phosphorylation and qPCR to assess gene expression. CAF induced a higher body mass gain in B1RKO compared to WT mice. CAF diet increased epididymal fat depot mass, hepatic fat infiltration and hepatic AKT phosphorylation in both genotypes. However, B1RKO mice presented lower glycemic response during GTT when fed with CAF, and a lower glucose decrease in the ITT. This higher resistance was overcomed with higher insulin secretion when stimulated by high glucose, resulting in higher glucose uptake in the GTT when submitted to CAF, despite lower insulin sensitivity. Islets from B1RKO delivered 4 times more insulin in 3-month-old mice than islets from WT. The higher insulin disposition index and high insulin delivery of B1RKO can explain the decreased glucose excursion during GTT. In conclusion, CAF increased the β-cell function in B1RKO mice, compensated by the diet-induced insulin resistance and resulting in a healthier glycemic response despite the higher weight gain.
Collapse
|
40
|
Moayedi K, Orandi S, Ebrahimi R, Tanhapour M, Moradi M, Abbastabar M, Golestani A. A novel approach to type 3 diabetes mechanism: The interplay between noncoding RNAs and insulin signaling pathway in Alzheimer's disease. J Cell Physiol 2022; 237:2838-2861. [PMID: 35580144 DOI: 10.1002/jcp.30779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 12/06/2022]
Abstract
Today, growing evidence indicates that patients with type 2 diabetes (T2D) are at a higher risk of developing Alzheimer's disease (AD). Indeed, AD as one of the main causes of dementia in people aged more than 65 years can be aggravated by insulin resistance (IR) and other metabolic risk factors related to T2D which are also linked to the function of the brain. Remarkably, a new term called "type 3 diabetes" has been suggested for those people who are diagnosed with AD while also showing the symptoms of IR and T2D. In this regard, the role of genetic and epigenetic changes associated with AD has been confirmed by many studies. On the other hand, it should be noted that the insulin signaling pathway is highly regulated by various mechanisms, including epigenetic factors. Among these, the role of noncoding RNAs (ncRNAs), including microRNAs and long noncoding RNAs has been comprehensively studied with respect to the pathology of AD and the most well-known underlying mechanisms. Nevertheless, the number of studies exploring the association between ncRNAs and the downstream targets of the insulin signaling pathway in the development of AD has notably increased in recent years. With this in view, the present study aimed to review the interplay between different ncRNAs and the insulin signaling pathway targets in the pathogenesis of AD to find a new approach in the field of combining biomarkers or therapeutic targets for this disease.
Collapse
Affiliation(s)
- Kiana Moayedi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Orandi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Tanhapour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Moradi
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Abbastabar
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Ye H, He Y, Zheng C, Wang F, Yang M, Lin J, Xu R, Zhang D. Type 2 Diabetes Complicated With Heart Failure: Research on Therapeutic Mechanism and Potential Drug Development Based on Insulin Signaling Pathway. Front Pharmacol 2022; 13:816588. [PMID: 35308248 PMCID: PMC8927800 DOI: 10.3389/fphar.2022.816588] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/31/2022] [Indexed: 01/16/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and heart failure (HF) are diseases characterized by high morbidity and mortality. They often occur simultaneously and increase the risk of each other. T2DM complicated with HF, as one of the most dangerous disease combinations in modern medicine, is more common in middle-aged and elderly people, making the treatment more difficult. At present, the combination of blood glucose control and anti-heart failure is a common therapy for patients with T2DM complicated with HF, but their effect is not ideal, and many hypoglycemic drugs have the risk of heart failure. Abnormal insulin signaling pathway, as a common pathogenic mechanism in T2DM and HF, could lead to pathological features such as insulin resistance (IR), myocardial energy metabolism disorders, and vascular endothelial disorders. The therapy based on the insulin signaling pathway may become a specific therapeutic target for T2DM patients with HF. Here, we reviewed the mechanisms and potential drugs of insulin signaling pathway in the treatment of T2DM complicated with HF, with a view to opening up a new perspective for the treatment of T2DM patients with HF and the research and development of new drugs.
Collapse
Affiliation(s)
- Hui Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanan He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Wang
- State Key Laboratory of Innovation Medicine and High Efficiency and Energy Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ming Yang
- State Key Laboratory of Innovation Medicine and High Efficiency and Energy Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Junzhi Lin
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Runchun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dingkun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
42
|
Shanak S, Bassalat N, Barghash A, Kadan S, Ardah M, Zaid H. Drug Discovery of Plausible Lead Natural Compounds That Target the Insulin Signaling Pathway: Bioinformatics Approaches. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2832889. [PMID: 35356248 PMCID: PMC8958086 DOI: 10.1155/2022/2832889] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/16/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
The growing smooth talk in the field of natural compounds is due to the ancient and current interest in herbal medicine and their potentially positive effects on health. Dozens of antidiabetic natural compounds were reported and tested in vivo, in silico, and in vitro. The role of these natural compounds, their actions on the insulin signaling pathway, and the stimulation of the glucose transporter-4 (GLUT4) insulin-responsive translocation to the plasma membrane (PM) are all crucial in the treatment of diabetes and insulin resistance. In this review, we collected and summarized a group of available in vivo and in vitro studies which targeted isolated phytochemicals with possible antidiabetic activity. Moreover, the in silico docking of natural compounds with some of the insulin signaling cascade key proteins is also summarized based on the current literature. In this review, hundreds of recent studies on pure natural compounds that alleviate type II diabetes mellitus (type II DM) were revised. We focused on natural compounds that could potentially regulate blood glucose and stimulate GLUT4 translocation through the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. On attempt to point out potential new natural antidiabetic compounds, this review also focuses on natural ingredients that were shown to interact with proteins in the insulin signaling pathway in silico, regardless of their in vitro/in vivo antidiabetic activity. We invite interested researchers to test these compounds as potential novel type II DM drugs and explore their therapeutic mechanisms.
Collapse
Affiliation(s)
- Siba Shanak
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Najlaa Bassalat
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
- Faculty of Medicine, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Ahmad Barghash
- Computer Science Department, German Jordanian University, Madaba Street. P.O. Box 35247, Amman 11180, Jordan
| | - Sleman Kadan
- Qasemi Research Center, Al-Qasemi Academic College, P.O Box 124, Baqa El-Gharbia 30100, Israel
| | - Mahmoud Ardah
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Hilal Zaid
- Faculty of Medicine, Arab American University, P.O Box 240, Jenin, State of Palestine
- Qasemi Research Center, Al-Qasemi Academic College, P.O Box 124, Baqa El-Gharbia 30100, Israel
| |
Collapse
|
43
|
Schwartzkopf CD, Hadcock JR, Liu G, Germano P, Roux J, Shea CM, Buys ES, Jones JE. Beneficial Metabolic Effects of Praliciguat, a Soluble Guanylate Cyclase Stimulator, in a Mouse Diet-Induced Obesity Model. Front Pharmacol 2022; 13:852080. [PMID: 35308230 PMCID: PMC8931041 DOI: 10.3389/fphar.2022.852080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Praliciguat is a soluble guanylate cyclase stimulator that elicits hemodynamic, anti-inflammatory, and antifibrotic effects in preclinical models of metabolic dysfunction. We assessed the metabolic effects of praliciguat in a mouse diet-induced obesity (DIO) model housed at thermoneutrality. At 6 weeks old, male C57BL/6N mice were either maintained on low-fat diet (LFD, lean mice) or placed on 60% high-fat diet (HFD, DIO mice). At 14 weeks old, the DIO mice were either maintained on HFD or switched to HFD with praliciguat (6-mg/kg). Day 28 samples were collected for biomarker analysis. In a second study under the same paradigm, indirect calorimetry was performed on days 8, 9, 20, 21, 32, and 33 and an oral lipid tolerance test (LTT) on day 38. Mice treated 28 days with praliciguat had lower levels of fasting plasma insulin, C-peptide, triglycerides, and HOMA-IR (homeostatic model assessment for insulin resistance) than DIO controls. In addition, energy expenditure was higher in praliciguat-treated than in DIO control mice on days 9, 20, 32, and 33; and day-38 triglycerides were lower. HFD-induced increases in gene expression of liver TNF-ɑ, lipoprotein lipase (Lpl), and patatin-like phospholipase domain-containing protein 3 (Pnpla3) in control DIO mice were attenuated in praliciguat-treated DIO mice. The positive metabolic effects observed in praliciguat-treated mice were associated with the restoration of liver PI3K (pAKT-Thr308) signaling, but not MAPK (pERK). In conclusion, praliciguat-treated DIO mice had increased energy utilization, improved insulin sensitivity, and lower plasma triglycerides. These results illustrate metabolic effects associated with praliciguat treatment in DIO mice.
Collapse
Affiliation(s)
| | | | - Guang Liu
- Cyclerion Therapeutics, Cambridge, MA, United States
| | - Peter Germano
- Cyclerion Therapeutics, Cambridge, MA, United States
| | | | | | | | - Juli E. Jones
- Cyclerion Therapeutics, Cambridge, MA, United States
- *Correspondence: Juli E. Jones,
| |
Collapse
|
44
|
Zhang C, Zhang D, Wang H, Lin Q, Li M, Yuan J, Gao G, Dong J. Hyperbaric oxygen treatment improves pancreatic β‑cell function and hepatic gluconeogenesis in STZ‑induced type‑2 diabetes mellitus model mice. Mol Med Rep 2022; 25:90. [PMID: 35039874 PMCID: PMC8809048 DOI: 10.3892/mmr.2022.12606] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/14/2021] [Indexed: 12/14/2022] Open
Abstract
Type‑2 diabetes mellitus (T2DM) causes several complications that affect the quality of life and life span of patients. Hyperbaric oxygen therapy (HBOT) has been used to successfully treat several diseases, including carbon monoxide poisoning, ischemia, infections and diabetic foot ulcer, and increases insulin sensitivity in T2DM. The present study aimed to determine the effect of HBOT on β‑cell function and hepatic gluconeogenesis in streptozotocin (STZ)‑induced type‑2 diabetic mice. To establish a T2DM model, 7‑week‑old male C57BL/6J mice were fed a high‑fat diet (HFD) and injected once daily with low‑dose STZ for 3 days after 1‑week HFD feeding. At the 14th week, HFD+HBOT and T2DM+HBOT groups received 1‑h HBOT (2 ATA; 100% pure O2) daily from 5:00 to 6:00 p.m. for 7 days. The HFD and T2DM groups were maintained under normobaric oxygen conditions and used as controls. During HBOT, the 12‑h nocturnal food intake and body weight were measured daily. Moreover, blood glucose was measured by using a tail vein prick and a glucometer. After the final HBO treatment, all mice were sacrificed to conduct molecular biology experiments. Fasting insulin levels of blood samples of sacrificed mice were measured by an ultrasensitive ELISA kit. Pancreas and liver tissues were stained with hematoxylin and eosin, while immunohistochemistry was performed to determine the effects of HBOT on insulin resistance. TUNEL was used to determine the effects of HBOT on β‑cell apoptosis, and immunoblotting was conducted to determine the β‑cell apoptosis pathway. HBOT notably reduced fasting blood glucose and improved insulin sensitivity in T2DM mice. After HBOT, β‑cell area and β‑cell mass in T2DM mice were significantly increased. HBOT significantly decreased the β‑cell apoptotic rate in T2DM mice via the pancreatic Bcl‑2/caspase‑3/poly(ADP‑ribose) polymerase (PARP) apoptosis pathway. Moreover, HBOT improved the morphology of the liver tissue and increased hepatic glycogen storage in T2DM mice. These findings suggested that HBOT ameliorated the insulin sensitivity of T2DM mice by decreasing the β‑cell apoptotic rate via the pancreatic Bcl‑2/caspase‑3/PARP apoptosis pathway.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Blood Glucose/metabolism
- Blotting, Western
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Fasting/blood
- Gluconeogenesis/physiology
- Glucose Tolerance Test/methods
- Humans
- Hyperbaric Oxygenation/methods
- Insulin/blood
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/metabolism
- Liver/metabolism
- Male
- Mice, Inbred C57BL
- Mice
Collapse
Affiliation(s)
- Caishun Zhang
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Di Zhang
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, Shandong 253023, P.R. China
| | - Haidan Wang
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Qian Lin
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Manwen Li
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Junhua Yuan
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Guangkai Gao
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
- Department of Hyperbaric Medicine, No. 971 Hospital of Chinese People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Jing Dong
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
- Physiology Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
45
|
Tóth ME, Sárközy M, Szűcs G, Dukay B, Hajdu P, Zvara Á, Puskás LG, Szebeni GJ, Ruppert Z, Csonka C, Kovács F, Kriston A, Horváth P, Kővári B, Cserni G, Csont T, Sántha M. Exercise training worsens cardiac performance in males but does not change ejection fraction and improves hypertrophy in females in a mouse model of metabolic syndrome. Biol Sex Differ 2022; 13:5. [PMID: 35101146 PMCID: PMC8805345 DOI: 10.1186/s13293-022-00414-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/05/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) refers to a cluster of co-existing cardio-metabolic risk factors, including visceral obesity, dyslipidemia, hyperglycemia with insulin resistance, and hypertension. As there is a close link between MetS and cardiovascular diseases, we aimed to investigate the sex-based differences in MetS-associated heart failure (HF) and cardiovascular response to regular exercise training (ET). METHODS High-fat diet-fed male and female APOB-100 transgenic (HFD/APOB-100, 3 months) mice were used as MetS models, and age- and sex-matched C57BL/6 wild-type mice on standard diet served as healthy controls (SD/WT). Both the SD/WT and HFD/APOB-100 mice were divided into sedentary and ET groups, the latter running on a treadmill (0.9 km/h) for 45 min 5 times per week for 7 months. At month 9, transthoracic echocardiography was performed to monitor cardiac function and morphology. At the termination of the experiment at month 10, blood was collected for serum low-density lipoprotein (LDL)- and high-density lipoprotein (HDL)-cholesterol measurements and homeostatic assessment model for insulin resistance (HOMA-IR) calculation. Cardiomyocyte hypertrophy and fibrosis were assessed by histology. Left ventricular expressions of selected genes associated with metabolism, inflammation, and stress response were investigated by qPCR. RESULTS Both HFD/APOB-100 males and females developed obesity and hypercholesterolemia; however, only males showed insulin resistance. ET did not change these metabolic parameters. HFD/APOB-100 males showed echocardiographic signs of mild HF with dilated ventricles and thinner walls, whereas females presented the beginning of left ventricular hypertrophy. In response to ET, SD/WT males developed increased left ventricular volumes, whereas females responded with physiologic hypertrophy. Exercise-trained HFD/APOB-100 males presented worsening HF with reduced ejection fraction; however, ET did not change the ejection fraction and reversed the echocardiographic signs of left ventricular hypertrophy in HFD/APOB-100 females. The left ventricular expression of the leptin receptor was higher in females than males in the SD/WT groups. Left ventricular expression levels of stress response-related genes were higher in the exercise-trained HFD/APOB-100 males and exercise-trained SD/WT females than exercise-trained SD/WT males. CONCLUSIONS HFD/APOB-100 mice showed sex-specific cardiovascular responses to MetS and ET; however, left ventricular gene expressions were similar between the groups except for leptin receptor and several stress response-related genes.
Collapse
Affiliation(s)
- Melinda E. Tóth
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720, Hungary. .,Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720, Hungary.
| | - Gergő Szűcs
- grid.9008.10000 0001 1016 9625MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720 Hungary ,grid.9008.10000 0001 1016 9625Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720 Hungary
| | - Brigitta Dukay
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Petra Hajdu
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Ágnes Zvara
- grid.418331.c0000 0001 2195 9606Laboratory of Functional Genomics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - László G. Puskás
- grid.418331.c0000 0001 2195 9606Laboratory of Functional Genomics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Gábor J. Szebeni
- grid.418331.c0000 0001 2195 9606Laboratory of Functional Genomics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Zsófia Ruppert
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,grid.9008.10000 0001 1016 9625Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Csaba Csonka
- grid.9008.10000 0001 1016 9625MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720 Hungary ,grid.9008.10000 0001 1016 9625Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720 Hungary
| | - Ferenc Kovács
- grid.481814.00000 0004 0479 9817Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726 Hungary
| | - András Kriston
- grid.481814.00000 0004 0479 9817Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Péter Horváth
- grid.481814.00000 0004 0479 9817Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726 Hungary ,grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Bence Kővári
- grid.9008.10000 0001 1016 9625Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720 Hungary
| | - Gábor Cserni
- grid.9008.10000 0001 1016 9625Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720 Hungary
| | - Tamás Csont
- grid.9008.10000 0001 1016 9625MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720 Hungary ,grid.9008.10000 0001 1016 9625Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720 Hungary
| | - Miklós Sántha
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| |
Collapse
|
46
|
Identification of Growth Factors, Cytokines and Mediators Regulated by Artemisia annua L. Polyphenols (pKAL) in HCT116 Colorectal Cancer Cells: TGF-β1 and NGF-β Attenuate pKAL-Induced Anticancer Effects via NF-κB p65 Upregulation. Int J Mol Sci 2022; 23:ijms23031598. [PMID: 35163520 PMCID: PMC8835737 DOI: 10.3390/ijms23031598] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 11/30/2022] Open
Abstract
The anticancer effects of natural phytochemicals are relevant to the modulation of cytokine signaling pathways in various cancer cells with stem-like properties as well as immune cells. The aim of this study was to elucidate a novel anticancer mechanism of Artemisia annua L. polyphenols (pKAL) involved in the regulation of growth factors, cytokines and mediators in stem-like HCT116 colorectal cancer cells. Through RayBiotech human L-1000 antibody array and bioinformatics analysis, we show here that pKAL-induced anticancer effects are associated with downregulation of growth factor and cytokine signaling proteins including TGFA, FGF16, PDGFC, CCL28, CXCR3, IRF6 and SMAD1. Notably, we found that TGF-β signaling proteins such as GDF10, ENG and TGFBR2 and well-known survival proteins such as NGF-β, VEGFD and insulin were significantly upregulated by pKAL. Moreover, the results of hematoxylin staining, cell viability assay and Western blot analysis demonstrated that TGF-β1 and NGF-β attenuated pKAL-induced anticancer effects by inhibiting pKAL-induced downregulation of caspase-8, NF-κB p65 and cyclin D1. These results suggest that certain survival mediators may be activated by pKAL through the TGF-β1 and NGF-β signaling pathways during pKAL-induced cell death and thus, strategies to inhibit the survival signaling are inevitably required for more effective anticancer effects of pKAL.
Collapse
|
47
|
Valença HDM, E Silva CP, de Brito Gitirana L, Valença SS, Lanzetti M. Beneficial effects of Ilex paraguariensis in the prevention of obesity-associated metabolic disorders in mice. Phytother Res 2022; 36:1032-1042. [PMID: 35028976 DOI: 10.1002/ptr.7377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/11/2022]
Abstract
Obesity is a chronic condition involving inflammation and oxidative stress that commonly predisposes affected individuals to develop metabolic disorders. We hypothesize that Ilex paraguariensis (IP) can modulate oxidative stress and inflammation underpinning metabolic disorders caused by obesity. C57BL/6 mice were fed a high-fat diet (HFD group) for 12 weeks. Concomitantly, some mice were treated with roasted IP (15 mg/ml - HFD + IP) or dimethyl fumarate (DMF) as a positive control (2 mg/ml - HFD + DMF). The control group received standard chow and water ad libitum. Histological analyses of fat tissue and liver, and quantification of mediators related to oxidative stress (Kelch-like ECH-associated protein 1/NF-E2-related factor 2, NADP(H) quinone oxidoreductase-1 [NQO1], heme oxygenase 1 [HO1], and superoxide dismutase) as well as metabolic profile blood biomarkers (glucose, leptin, resistin, high-density lipoproteins [HDLs], and triglycerides) were performed. Metabolic disorders were prevented in mice treated with IP, as evidenced by the observation that glucose, HDL, and resistin levels were similar to those assessed in the control group. Morphological analyses showed that both IP and DMF treatments prevented hepatic steatosis and adipocyte hypertrophy in visceral adipose tissue. Finally, although the antioxidant response stimulated by IP was quite limited, significant effects were found on NQO1 and HO1 expression. In conclusion, IP has promising preventative effects on the development of metabolic disorders caused by obesity.
Collapse
Affiliation(s)
- Helber da Maia Valença
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cyntia Pecli E Silva
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lycia de Brito Gitirana
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Samuel Santos Valença
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Manuella Lanzetti
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
48
|
LUO W, Liu F, QI X, DONG G. Research progress of konjac dietary fibre in the prevention and treatment of diabetes. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.23322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Wanyu LUO
- Shandong Traditional Chinese Medicine University, China; Qingdao Chengyang District Yuhe Tang Chinese Medicine clinic, China
| | - Fanghua Liu
- Qingdao Chengyang District Yuhe Tang Chinese Medicine clinic, China
| | - Xin QI
- Guang’anmen Hospital of China Academy of Chinese Medical Sciences, China
| | | |
Collapse
|
49
|
He Y, Ding F, Yin M, Zhang H, Hou L, Cui T, Xu J, Yue J, Zheng Q. High Serum AST/ALT Ratio and Low Serum INS*PA Product Are Risk Factors and Can Diagnose Sarcopenia in Middle-Aged and Older Adults. Front Endocrinol (Lausanne) 2022; 13:843610. [PMID: 35370985 PMCID: PMC8971564 DOI: 10.3389/fendo.2022.843610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/14/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Sarcopenia is an age-related clinical condition and associated with an increased risk of adverse outcomes. However, to date, there is no global standard for the diagnosis of sarcopenia, and fewer serum biomarkers have been suggested for the diagnosis of sarcopenia. It is, thus, important that sarcopenia-related serological diagnostic markers be explored. The present study was based on the Asian Working Group on Sarcopenia 2019 (AWGS 2019) criteria to assess whether aspartate aminotransferase/alanine aminotransferase (AST/ALT) ratio and fasting insulin*prealbumin (INS*PA) product are diagnostic markers associated with sarcopenia in various ethnic groups in western China. METHODS This cross-sectional study included 4,099 adults (1,471 men and 2,628 women) from the West China Health and Aging Trend (WCHAT) study. The value of serum biomarkers was based on laboratory data. The accompanying metabolic disorders and the associated parameters were evaluated. Logistic regression analysis was used to explore the association between markers and sarcopenia. Receiver operating characteristic curve (ROC) analysis was used to evaluate the diagnostic efficacy of the test in differentiating sarcopenia. RESULTS Binary regression analysis showed that high serum AST/ALT (OR = 2.247) and adrenal cortisol (PTC, OR = 1.511), low serum INS*PA (OR = 2.970), free triiodothyronine (FT3, OR = 1.313), 25-OH-VitD (VitD, in male participants, OR = 1.817), and diastolic blood pressure (DBP, in female subjects, OR = 1.250) were independent risk factors for sarcopenia (P < 0.05). AST/ALT and INS*PA were not affected by metabolic factors and had better diagnostic efficacy for sarcopenia. The AUC of the INS*PA was the highest (0.705, 0.706, and 0.701, respectively, P < 0.05), followed by that of the AST/ALT (0.680, 0.675, and 0.695, respectively, P < 0.05). The AUC of the AST/ALT/(INS*PA)*10,000 used to diagnose sarcopenia was 0.727. CONCLUSION Among middle-aged and older adults of multiple ethnicities in western China, we found that higher AST/ALT and lower INS*PA levels are associated with an increased prevalence of sarcopenia. Since these serum biomarkers are inexpensive and can be obtained easily from biochemical routine, regular follow-up of AST/ALT and INS*PA may be an effective strategy in sarcopenia screening and management.
Collapse
Affiliation(s)
- Yong He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Fing Ding
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Mengting Yin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lisha Hou
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Cui
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Jinfeng Xu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Jirong Yue
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qin Zheng, ; Jirong Yue,
| | - Qin Zheng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qin Zheng, ; Jirong Yue,
| |
Collapse
|
50
|
Wang H, Zhang J, Pu Y, Qin S, Liu H, Tian Y, Tang Z. Comparison of different insulin resistance surrogates to predict hyperuricemia among U.S. non-diabetic adults. Front Endocrinol (Lausanne) 2022; 13:1028167. [PMID: 36589794 PMCID: PMC9797589 DOI: 10.3389/fendo.2022.1028167] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Although it has been well-acknowledged that insulin resistance (IR) plays a critical role in the development of hyperuricemia (HU), specific relationship between IR and HU in non-diabetic patients remains rarely studied, and there is still no large-scale research regarding this issue. This study aims to explore the association between triglyceride glucose (TyG), TyG with body mass index (TyG-BMI), the ratio of triglycerides divided by high-density lipoprotein cholesterol (TG/HDL-C), metabolic score for insulin resistance (METS-IR), and the risk of HU in non-diabetic patients in The United States of America. PATIENTS AND METHODS Data from the National Health and Nutrition Examination Survey (NHANES) enrolling a representative population aged ≥18-year-old were included to calculate these four indexes. Logistic regression analysis was applied to describe their associations and calculate odds ratios (OR) while the Receiver Operating Characteristic curve was utilized to assess the prediction ability of these four indexes. RESULTS A total of 7,743 people (3,806 males and 3,937 females, mean age: 45.17 ± 17.10 years old) were included in this study, among whom 32.18% suffered from HU. After adjustment for sex, age, ethnicity, education background, smoking status, drinking status, systolic blood pressure (SBP), diastolic blood pressure (DBP), metabolic equivalent values (METs), total cholesterol, low-density lipoprotein cholesterol, and estimated glomerular filtration rate, it showed that all four indexes were closely related to HU. Compared with the lowest quartile, OR of the highest quartile of these four indicators for HU were as following respectively: TyG: 5.61 (95% CI: 4.29-7.32); TyG-BMI: 7.15 (95% CI: 5.56-9.20); TG/HDL-C: 4.42 (95% CI: 3.49-5.60); METS-IR: 7.84 (95% CI: 6.07-10.13). TyG, TyG-BMI, TG/HDL-C and METS-IR had moderate discrimination ability for HU, with an AUC value of 0.66 (95% CI: 0.65-0.68), 0.67 (95% CI: 0.65-0.68), 0.68 (95% CI: 0.67-0.69) and 0.68 (95% CI: 0.66-0.69) respectively. Each index showed better prediction ability for HU risk in females than in males. CONCLUSION It was found that the risk of HU was positively associated with the elevation of TyG, TyG-BMI, TG/HDL-C and METS-IR in a large-scale population of U.S., and TyG-BMI and METS-IR have a better ability to identify HU in both genders.
Collapse
Affiliation(s)
- Hao Wang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jia Zhang
- Department of Cardiology, Shanghai Seventh People’s Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuzhu Pu
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Shengmei Qin
- Medicine & Health Science of Huangshang University, Guangzhou, China
| | - Huan Liu
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yongming Tian
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zhihong Tang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Zhihong Tang,
| |
Collapse
|