1
|
El-Eshmawy MM, Mahsoub N, Elsehely I. Serum total bilirubin is a risk factor of metabolic syndrome and its components in obese Egyptians. Porto Biomed J 2024; 9:274. [PMID: 39563980 PMCID: PMC11573332 DOI: 10.1097/j.pbj.0000000000000275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/26/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Background/Aim The link between serum total bilirubin and metabolic syndrome and its components has been previously proposed. However, it is unknown whether total bilirubin is a risk factor of metabolic syndrome and its components in obese Egyptians. Therefore, this study was conducted to clarify the association of total bilirubin levels with metabolic syndrome and its components in obese Egyptians. Methods A total of 200 adults with obesity were enrolled in this study. Obese participants were evaluated for metabolic syndrome; there were 92 obese participants with metabolic syndrome and 108 obese participants without metabolic syndrome. Anthropometric measurements, fasting blood glucose (FBG), fasting insulin, homeostasis model assessment of insulin resistance (HOMA-IR), HOMA-β (%), lipid profile, uric acid, alanine aminotransferase, aspartate aminotransferase, and serum total bilirubin were assessed. Results Total bilirubin was significantly lower in obese participants with metabolic syndrome than in those without metabolic syndrome. Compared with middle bilirubin tertile, high and low bilirubin tertiles were independently associated with metabolic syndrome. Regarding metabolic syndrome components, a significant positive association between low bilirubin tertile and hypertension was found independent of the all studied confounding factors, whereas the association of total bilirubin level with waist circumference (WC), FBG, high-density lipoprotein cholesterol, and triglycerides was dependent on body mass index (BMI), HOMA-IR, and high sensitive C-reactive protein (hs-CRP). Conclusion Total bilirubin is an independent risk factor of metabolic syndrome in obese Egyptians. We have found an independent association between high bilirubin level and reduced risk of metabolic syndrome, whereas low bilirubin level was associated with increased risk of metabolic syndrome. Bilirubin is also independently associated with hypertension, but its association with other components of metabolic syndrome is mainly dependent on BMI, HOMA-IR, and hs-CRP.
Collapse
Affiliation(s)
- Mervat M El-Eshmawy
- Internal Medicine Department, Mansoura Specialized Medical Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nancy Mahsoub
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ibrahim Elsehely
- Internal Medicine Department, Mansoura Specialized Medical Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
2
|
Chen F, Cao LH, Ma FY, Zeng LL, He JR. Development and validation of a predictive model for severe white matter hyperintensity with obesity. Front Aging Neurosci 2024; 16:1404756. [PMID: 38887608 PMCID: PMC11180876 DOI: 10.3389/fnagi.2024.1404756] [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: 03/21/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Purpose The purpose of the present study was to identify predictors of severe white matter hyperintensity (WMH) with obesity (SWO), and to build a prediction model for screening obese people with severe WMH without Nuclear Magnetic Resonance Imaging (MRI) examination. Patients subjects and methods From September 2020 to October 2021, 650 patients with WMH were recruited consecutively. The subjects were divided into two groups, SWO group and non-SWO group. Univariate and Logistic regression analysis were was applied to explore the potential predictors of SWO. The Youden index method was adopted to determine the best cut-off value in the establishment of the prediction model of SWO. Each parameter had two options, low and high. The score table of the prediction model and nomogram based on the logistic regression were constructed. Of the 650 subjects, 487 subjects (75%) were randomly assigned to the training group and 163 subjects (25%) to the validation group. By resampling the area under the curve (AUC) of the subject's operating characteristics and calibration curves 1,000 times, nomogram performance was verified. A decision curve analysis (DCA) was used to evaluate the nomogram's clinical usefulness. By resampling the area under the curve (AUC) of the subject's operating characteristics and calibration curves 1,000 times, nomogram performance was verified. A decision curve analysis (DCA) was used to evaluate the nomogram's clinical usefulness. Results Logistic regression demonstrated that hypertension, uric acid (UA), complement 3 (C3) and Interleukin 8 (IL-8) were independent risk factors for SWO. Hypertension, UA, C3, IL-8, folic acid (FA), fasting C-peptide (FCP) and eosinophil could be used to predict the occurrence of SWO in the prediction models, with a good diagnostic performance, Areas Under Curves (AUC) of Total score was 0.823 (95% CI: 0.760-0.885, p < 0.001), sensitivity of 60.0%, specificity of 91.4%. In the development group, the nomogram's AUC (C statistic) was 0.829 (95% CI: 0.760-0.899), while in the validation group, it was 0.835 (95% CI: 0.696, 0.975). In both the development and validation groups, the calibration curves following 1,000 bootstraps showed a satisfactory fit between the observed and predicted probabilities. DCA showed that the nomogram had great clinical utility. Conclusion Hypertension, UA, C3, IL-8, FA, FCP and eosinophil models had the potential to predict the incidence of SWO. When the total score of the model exceeded 9 points, the risk of SWO would increase significantly, and the nomogram enabled visualization of the patient's WMH risk. The application prospect of our models mainly lied in the convenient screening of SWO without MRI examination in order to detect SWO and control the WMH hazards early.
Collapse
Affiliation(s)
- Fu Chen
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Medicine, Yinhang Community Health Centre, Shanghai, China
| | - Lin-Hao Cao
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei-Yue Ma
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Li Zeng
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Rong He
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Jussila A, Zhang B, Kirti S, Atit R. Tissue fibrosis associated depletion of lipid-filled cells. Exp Dermatol 2024; 33:e15054. [PMID: 38519432 PMCID: PMC10977660 DOI: 10.1111/exd.15054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/24/2024]
Abstract
Fibrosis is primarily described as the deposition of excessive extracellular matrix, but in many tissues it also involves a loss of lipid or lipid-filled cells. Lipid-filled cells are critical to tissue function and integrity in many tissues including the skin and lungs. Thus, loss or depletion of lipid-filled cells during fibrogenesis, has implications for tissue function. In some contexts, lipid-filled cells can impact ECM composition and stability, highlighting their importance in fibrotic transformation. Recent papers in fibrosis address this newly recognized fibrotic lipodystrophy phenomenon. Even in disparate tissues, common mechanisms are emerging to explain fibrotic lipodystrophy. These findings have implications for fibrosis in tissues composed of fibroblast and lipid-filled cell populations such as skin, lung, and liver. In this review, we will discuss the roles of lipid-containing cells, their reduction/loss during fibrotic transformation, and the mechanisms of that loss in the skin and lungs.
Collapse
Affiliation(s)
- Anna Jussila
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brian Zhang
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sakin Kirti
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Radhika Atit
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
4
|
Sun Y, Xu H, Li J, Peng M, Jia Z, Kong L, Zhang X, Shao S, Zhang W, Wang W. Genome-wide survey identifies TNNI2 as a target of KLF7 that inhibits chicken adipogenesis via downregulating FABP4. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194899. [PMID: 36410687 DOI: 10.1016/j.bbagrm.2022.194899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/26/2022] [Accepted: 11/12/2022] [Indexed: 11/19/2022]
Abstract
Krüppel-like factor 7 (KLF7) negatively regulates adipocyte differentiation; however, the mechanism underlying its activity in mammals and birds remains poorly understood. To identify genome-wide KLF7-binding motifs in preadipocytes, we conducted a chromatin immunoprecipitation-sequencing analysis of immortalized chicken preadipocytes (ICP2), which revealed 11,063 specific binding sites. Intergenic binding site analysis showed that KLF7 regulates several novel factors whose functions in chicken and mammal adipogenesis are underexplored. We identified a novel regulator, troponin I2 (TNNI2), which is positively regulated by KLF7. TNNI2 is downregulated during preadipocyte differentiation and acts as an adipogenic repressor at least in part by repressing FABP4 promoter activity. In conclusion, we demonstrated that KLF7 functions through cis-regulation of TNNI2, which inhibits adipogenesis. Our findings not only provide the first genome-wide picture of KLF7 associations in preadipocytes but also identify a novel function of TNNI2.
Collapse
Affiliation(s)
- Yingning Sun
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China.
| | - Hu Xu
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Jinwei Li
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Min Peng
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Ziqiu Jia
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Lingzhe Kong
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Xin Zhang
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Shuli Shao
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Weiwei Zhang
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| | - Weiyu Wang
- College of Life Science and Agriculture Forestry, Qiqihar University, Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas, Qiqihar, Heilongjiang 161000, China
| |
Collapse
|
5
|
Feng Z, Chen J, Chen C, Feng L, Wang R, Zhu J, Lou R, Liu J, Ye Y, Lin L. Bioactivity-based molecular networking-guided identification of guttiferone J from Garcinia cambogia as an anti-obesity candidate. Br J Pharmacol 2023; 180:589-608. [PMID: 36321884 DOI: 10.1111/bph.15979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 10/03/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND AND PURPOSE Pharmacological intervention to induce browning of white adipose tissue provides a promising anti-obesity therapy. The fruits of Garcinia cambogia (Clusiaceae) have been widely applied to manage body weight; however, the chemical principles remain unclear. The current study aims to discover browning inducers from the fruits of G. cambogia and investigate the underlying mechanisms. EXPERIMENTAL APPROACH The bioactivity-based molecular networking and Oil Red O staining on 3T3-L1 and C3H10T1/2 adipocytes were applied for guided isolation. High-fat diet-induced obese mice were recruited to evaluate the anti-obesity activity. KEY RESULTS The bioactivity-based molecular networking-guided isolation yielded several polycyclic polyprenylated acylphloroglucinols from the fruits of G. cambogia with lipid-lowering effect in adipocytes, including guttiferone J (GOJ), garcinol and 14-deoxygarcinol. As the most potent one, GOJ (10 μM) reduced lipid accumulation by 70% and 76% in 3T3-L1 and C3H10T1/2 adipocytes, respectively. Furthermore, GOJ (2.5-10 μM) increased the expression of the deacetylase sirtuin 3 (SIRT3) and activated it, which, in turn, reduced the acetylation level of PPARγ coactivator-1α to boost mitochondrial biogenesis and promoted uncoupling protein 1 expression to enhance thermogenesis, resulting in browning of adipocytes. In high-fat diet-induced-obese mice, GOJ (10 and 20 mg·kg-1 ·day-1 for 12 weeks) protected against adiposity, hyperlipidaemia, insulin resistance and liver lipotoxicity, through boosting SIRT3-mediated browning of inguinal adipose tissue. CONCLUSION AND IMPLICATIONS GOJ represents a new scaffold of thermogenic inducer, which is responsible for the anti-obesity property of G. cambogia and can be further developed as a candidate for treating obesity and its related disorders.
Collapse
Affiliation(s)
- Zheling Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jiali Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Cheng Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Lu Feng
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Rui Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jianzhong Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ruohan Lou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jia Liu
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yang Ye
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.,Department of Pharmaceutical Sciences and Technology, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
6
|
Wang G, Song A, Bae M, Wang QA. Adipose Tissue Plasticity in Aging. Compr Physiol 2022; 12:4119-4132. [PMID: 36214190 DOI: 10.1002/cphy.c220005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
As a dynamic endocrine organ, white adipose tissue (WAT) stores lipids and plays a critical role in maintaining whole-body energy homeostasis and insulin sensitivity. A large group of the population over 65 years old suffer from increased WAT mass, especially in the visceral location. Visceral adiposity accelerates aging through promoting age-associated chronic conditions, significantly shortening life expectancy. Unlike WAT, brown adipose tissue (BAT) functions as an effective energy sink that burns and disposes of excess lipids and glucose upon activation of thermogenesis. Unfortunately, the thermogenic activity of BAT declines during aging. New appreciation of cellular and functional remodeling of WAT and BAT during aging has emerged in recent years. Efforts are underway to explore the potential underlying mechanisms behind these age-associated alterations in WAT and BAT and the impact of these alterations on whole-body metabolism. Lastly, it is intriguing to translate our knowledge obtained from animal models to the clinic to prevent and treat age-associated metabolic disorders. © 2022 American Physiological Society. Compr Physiol 12: 4119-4132, 2022.
Collapse
Affiliation(s)
- Guan Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Anying Song
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Marie Bae
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Qiong A Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA.,Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
7
|
Nutraceutical Prevention of Diabetic Complications—Focus on Dicarbonyl and Oxidative Stress. Curr Issues Mol Biol 2022; 44:4314-4338. [PMID: 36135209 PMCID: PMC9498143 DOI: 10.3390/cimb44090297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/25/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Oxidative and dicarbonyl stress, driven by excess accumulation of glycolytic intermediates in cells that are highly permeable to glucose in the absence of effective insulin activity, appear to be the chief mediators of the complications of diabetes. The most pathogenically significant dicarbonyl stress reflects spontaneous dephosphorylation of glycolytic triose phosphates, giving rise to highly reactive methylglyoxal. This compound can be converted to harmless lactate by the sequential activity of glyoxalase I and II, employing glutathione as a catalyst. The transcription of glyoxalase I, rate-limiting for this process, is promoted by Nrf2, which can be activated by nutraceutical phase 2 inducers such as lipoic acid and sulforaphane. In cells exposed to hyperglycemia, glycine somehow up-regulates Nrf2 activity. Zinc can likewise promote glyoxalase I transcription, via activation of the metal-responsive transcription factor (MTF) that binds to the glyoxalase promoter. Induction of glyoxalase I and metallothionein may explain the protective impact of zinc in rodent models of diabetic complications. With respect to the contribution of oxidative stress to diabetic complications, promoters of mitophagy and mitochondrial biogenesis, UCP2 inducers, inhibitors of NAPDH oxidase, recouplers of eNOS, glutathione precursors, membrane oxidant scavengers, Nrf2 activators, and correction of diabetic thiamine deficiency should help to quell this.
Collapse
|
8
|
Chou CL, Li CH, Fang TC. Benefits of Valsartan and Amlodipine in Lipolysis through PU.1 Inhibition in Fructose-Induced Adiposity. Nutrients 2022; 14:nu14183759. [PMID: 36145135 PMCID: PMC9502698 DOI: 10.3390/nu14183759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
High fructose intake has been implicated in obesity and metabolic syndrome, which are related to increased cardiovascular mortality. However, few studies have experimentally examined the role of renin–angiotensin system blockers and calcium channel blockers (CCB) in obesity. We investigated the effects of valsartan (an angiotensin II receptor blocker) and amlodipine (a CCB) on lipolysis through the potential mechanism of PU.1 inhibition. We observed that high fructose concentrations significantly increased adipose size and triglyceride, monoacylglycerol lipase, adipose triglyceride lipase, and stearoyl-CoA desaturase-1 (SCD1), activating transcription factor 3 and PU.1 levels in adipocytes in vitro. Subsequently, PU.1 inhibitor treatment was able to reduce triglyceride, SCD1, and PU.1 levels. In addition, elevated levels of triglyceride and PU.1, stimulated by a high fructose concentration, decreased with valsartan and amlodipine treatment. Overall, these findings suggest that high fructose concentrations cause triacylglycerol storage in adipocytes through PU.1-mediated activation. Furthermore, valsartan and amlodipine treatment reduced triacylglycerol storage in adipocytes by inhibiting PU.1 activation in high fructose concentrations in vitro. Thus, the benefits of valsartan and amlodipine in lipolysis may be through PU.1 inhibition in fructose-induced adiposity, and PU.1 inhibition might have a potential therapeutic role in lipolysis in fructose-induced obesity.
Collapse
Affiliation(s)
- Chu-Lin Chou
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City 320, Taiwan
| | - Ching-Hao Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Te-Chao Fang
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: ; Tel.: +886-2-2737-2181
| |
Collapse
|
9
|
Chen C, Ren YM, Zhu JZ, Chen JL, Feng ZL, Zhang T, Ye Y, Lin LG. Ainsliadimer C, a disesquiterpenoid isolated from Ainsliaea macrocephala, ameliorates inflammatory responses in adipose tissue via Sirtuin 1-NLRP3 inflammasome axis. Acta Pharmacol Sin 2022; 43:1780-1792. [PMID: 34789920 PMCID: PMC9253034 DOI: 10.1038/s41401-021-00797-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022] Open
Abstract
Interleukin-1β (IL-1β), a key pro-inflammatory cytokine, is majorly produced by macrophages through NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, which has been identified as the culprit to deteriorate the inflammatory crosstalk between macrophages and adipocytes. Ainsliadimer C (AC) is a disesquiterpenoid isolated from Ainsliaea macrocephala. In the current study, we investigated the effects of AC on adipose tissue inflammation in co-culture of macrophages and adipocytes in vitro as well as in LPS-treated mice in vivo. We showed that AC (20-80 µM) dose-dependently inhibited the secretion of IL-1β from LPS plus ATP-stimulated THP-1 macrophages by inhibiting the activation of NLRP3 inflammasome. Furthermore, we found that AC treatment activated NAD+-dependent deacetylase Sirtuin 1 (SIRT1), resulting in reduced acetylation level of NLRP3. Molecular modeling analysis revealed that binding of AC to sirtuin-activating compound-binding domain increased the affinity of the substrate to the catalytic domain of SIRT1. Moreover, AC (80 µM) significantly attenuated macrophage-conditioned medium-induced inflammatory responses in 3T3-L1 adipocytes. In LPS-induced acute inflammatory mice, administration of AC (20, 60 mg·kg-1·d-1, ip) for 5 days significantly suppressed the pro-inflammatory cytokine levels in serum and epididymal white adipose tissue (eWAT), attenuated macrophage infiltration into eWAT, and mitigated adipose tissue inflammation. The beneficial effects of AC were blocked by co-administration of a selective SIRT1 inhibitor EX-527 (10 mg·kg-1·d-1). Taken together, AC suppresses NLRP3-mediated IL-1β secretion through activating SIRT1, leading to attenuated inflammation in macrophages and adipose tissue, which might be a candidate to treat obesity-associated metabolic diseases.
Collapse
Affiliation(s)
- Cheng Chen
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Yong-mei Ren
- grid.9227.e0000000119573309State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jian-zhong Zhu
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Jia-li Chen
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Zhe-ling Feng
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Tian Zhang
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Yang Ye
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Li-gen Lin
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| |
Collapse
|
10
|
Chen KY, De Angulo A, Guo X, More A, Ochsner SA, Lopez E, Saul D, Pang W, Sun Y, McKenna NJ, Tong Q. Adipocyte-Specific Ablation of PU.1 Promotes Energy Expenditure and Ameliorates Metabolic Syndrome in Aging Mice. FRONTIERS IN AGING 2022; 2:803482. [PMID: 35822007 PMCID: PMC9261351 DOI: 10.3389/fragi.2021.803482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/13/2021] [Indexed: 12/03/2022]
Abstract
Objective: Although PU.1/Spi1 is known as a master regulator for macrophage development and function, we have reported previously that it is also expressed in adipocytes and is transcriptionally induced in obesity. Here, we investigated the role of adipocyte PU.1 in the development of the age-associated metabolic syndrome. Methods: We generated mice with adipocyte-specific PU.1 knockout, assessed metabolic changes in young and older adult PU.1fl/fl (control) and AdipoqCre PU.1fl/fl (aPU.1KO) mice, including body weight, body composition, energy expenditure, and glucose homeostasis. We also performed transcriptional analyses using RNA-Sequencing of adipocytes from these mice. Results: aPU.1KO mice have elevated energy expenditure at a young age and decreased adiposity and increased insulin sensitivity in later life. Corroborating these observations, transcriptional network analysis indicated the existence of validated, adipocyte PU.1-modulated regulatory hubs that direct inflammatory and thermogenic gene expression programs. Conclusion: Our data provide evidence for a previously uncharacterized role of PU.1 in the development of age-associated obesity and insulin resistance.
Collapse
Affiliation(s)
- Ke Yun Chen
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Alejandra De Angulo
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Xin Guo
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Aditya More
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Eduardo Lopez
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - David Saul
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Weijun Pang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Northwestern University of Agriculture and Forestry, Yangling, China
| | - Yuxiang Sun
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Neil J. McKenna, ; Qiang Tong,
| | - Qiang Tong
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Huffington Center on Aging, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Huffington Center on Aging, Houston, TX, United States
- *Correspondence: Neil J. McKenna, ; Qiang Tong,
| |
Collapse
|
11
|
Liu G, Luo S, Lei Y, Wu J, Huang Z, Wang K, Yang P, Huang X. A nine-hub-gene signature of metabolic syndrome identified using machine learning algorithms and integrated bioinformatics. Bioengineered 2021; 12:5727-5738. [PMID: 34516309 PMCID: PMC8806918 DOI: 10.1080/21655979.2021.1968249] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Early risk assessments and interventions for metabolic syndrome (MetS) are limited because of a lack of effective biomarkers. In the present study, several candidate genes were selected as a blood-based transcriptomic signature for MetS. We collected so far the largest MetS-associated peripheral blood high-throughput transcriptomics data and put forward a novel feature selection strategy by combining weighted gene co-expression network analysis, protein-protein interaction network analysis, LASSO regression and random forest approaches. Two gene modules and 51 hub genes as well as a 9-hub-gene signature associated with metabolic syndrome were identified. Then, based on this 9-hub-gene signature, we performed logistic analysis and subsequently established a web nomogram calculator for metabolic syndrome risk (https://xjtulgz.shinyapps.io/DynNomapp/). This 9-hub-gene signature showed excellent classification and calibration performance (AUC = 0.968 in training set, AUC = 0.883 in internal validation set, AUC = 0.861 in external validation set) as well as ideal potential clinical benefit.
Collapse
Affiliation(s)
- Guanzhi Liu
- Bone and Joint Surgery Center, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Sen Luo
- Bone and Joint Surgery Center, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yutian Lei
- Bone and Joint Surgery Center, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jianhua Wu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhuo Huang
- Bone and Joint Surgery Center, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kunzheng Wang
- Bone and Joint Surgery Center, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Pei Yang
- Bone and Joint Surgery Center, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xin Huang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
12
|
Annie-Mathew AS, Prem-Santhosh S, Jayasuriya R, Ganesh G, Ramkumar KM, Sarada DVL. The pivotal role of Nrf2 activators in adipocyte biology. Pharmacol Res 2021; 173:105853. [PMID: 34455076 DOI: 10.1016/j.phrs.2021.105853] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/07/2023]
Abstract
Adipose tissue is instrumental in maintaining metabolic homeostasis by regulating energy storage in the form of triglycerides. In the case of over-nutrition, adipocytes favorably regulate lipogenesis over lipolysis and accumulate excess triglycerides, resulting in increased adipose tissue mass. An abnormal increase in hypertrophic adipocytes is associated with chronic complications such as insulin resistance, obesity, diabetes, atherosclerosis and nonalcoholic fatty liver disease. Experimental studies indicate the occurrence of oxidative stress in the pathogenesis of obesity. A common underlying link between increasing adipose tissue mass and oxidative stress is the Nuclear Factor Erythroid 2-related factor 2 (Nrf2), Keap1-Nrf2-ARE signaling, which plays an indispensable role in metabolic homeostasis by regulating oxidative and inflammatory responses. Additionally, Nrf2 also activates CCAAT/enhancer-binding protein α, (C/EBP-α), C/EBP-β and peroxisome proliferator-activated receptor γ (PPARγ) the crucial pro-adipogenic factors that promote de novo adipogenesis. Hence, at the forefront of research is the quest for prospecting novel compounds to modulate Nrf2 activity in the context of adipogenesis and obesity. This review summarizes the molecular mechanism behind the activation of the Keap1-Nrf2-ARE signaling network and the role of Nrf2 activators in adipocyte pathophysiology.
Collapse
Affiliation(s)
- A S Annie-Mathew
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Subramanian Prem-Santhosh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Ravichandran Jayasuriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Goutham Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - D V L Sarada
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
13
|
An automated framework for efficiently designing deep convolutional neural networks in genomics. NAT MACH INTELL 2021. [DOI: 10.1038/s42256-021-00316-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
14
|
Andrade S, Morais T, Sandovici I, Seabra AL, Constância M, Monteiro MP. Adipose Tissue Epigenetic Profile in Obesity-Related Dysglycemia - A Systematic Review. Front Endocrinol (Lausanne) 2021; 12:681649. [PMID: 34290669 PMCID: PMC8288106 DOI: 10.3389/fendo.2021.681649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/26/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Obesity is a major risk factor for dysglycemic disorders, including type 2 diabetes (T2D). However, there is wide phenotypic variation in metabolic profiles. Tissue-specific epigenetic modifications could be partially accountable for the observed phenotypic variability. SCOPE The aim of this systematic review was to summarize the available data on epigenetic signatures in human adipose tissue (AT) that characterize overweight or obesity-related insulin resistance (IR) and dysglycemia states and to identify potential underlying mechanisms through the use of unbiased bioinformatics approaches. METHODS Original data published in the last decade concerning the comparison of epigenetic marks in human AT of individuals with metabolically unhealthy overweight/obesity (MUHO) versus normal weight individuals or individuals with metabolically healthy overweight/obesity (MHO) was assessed. Furthermore, association of these epigenetic marks with IR/dysglycemic traits, including T2D, was compiled. RESULTS We catalogued more than two thousand differentially methylated regions (DMRs; above the cut-off of 5%) in the AT of individuals with MUHO compared to individuals with MHO. These DNA methylation changes were less likely to occur around the promoter regions and were enriched at loci implicated in intracellular signaling (signal transduction mediated by small GTPases, ERK1/2 signaling and intracellular trafficking). We also identified a network of seven transcription factors that may play an important role in targeting DNA methylation changes to specific genes in the AT of subjects with MUHO, contributing to the pathogeny of obesity-related IR/T2D. Furthermore, we found differentially methylated CpG sites at 8 genes that were present in AT and whole blood, suggesting that DMRs in whole blood could be potentially used as accessible biomarkers of MUHO. CONCLUSIONS The overall evidence linking epigenetic alterations in key tissues such AT to metabolic complications in human obesity is still very limited, highlighting the need for further studies, particularly those focusing on epigenetic marks other than DNA methylation. Our initial analysis suggests that DNA methylation patterns can potentially discriminate between MUHO from MHO and provide new clues into why some people with obesity are less susceptible to dysglycemia. Identifying AT-specific epigenetic targets could also lead to novel approaches to modify the progression of individuals with obesity towards metabolic disease. SYSTEMATIC REVIEW REGISTRATION PROSPERO, identifier CRD42021227237.
Collapse
Affiliation(s)
- Sara Andrade
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Tiago Morais
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Ionel Sandovici
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Alexandre L. Seabra
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Miguel Constância
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- National Institute of Health Research, Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Mariana P. Monteiro
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- *Correspondence: Mariana P. Monteiro,
| |
Collapse
|
15
|
Implications of metabolism-driven myeloid dysfunctions in cancer therapy. Cell Mol Immunol 2020; 18:829-841. [PMID: 33077904 PMCID: PMC7570408 DOI: 10.1038/s41423-020-00556-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
Immune homeostasis is maintained by an adequate balance of myeloid and lymphoid responses. In chronic inflammatory states, including cancer, this balance is lost due to dramatic expansion of myeloid progenitors that fail to mature to functional inflammatory neutrophils, macrophages, and dendritic cells (DCs), thus giving rise to a decline in the antitumor effector lymphoid response. Cancer-related inflammation orchestrates the production of hematopoietic growth factors and cytokines that perpetuate recruitment and activation of myeloid precursors, resulting in unresolved and chronic inflammation. This pathologic inflammation creates profound alterations in the intrinsic cellular metabolism of the myeloid progenitor pool, which is amplified by competition for essential nutrients and by hypoxia-induced metabolic rewiring at the tumor site. Therefore, persistent myelopoiesis and metabolic dysfunctions contribute to the development of cancer, as well as to the severity of a broad range of diseases, including metabolic syndrome and autoimmune and infectious diseases. The aims of this review are to (1) define the metabolic networks implicated in aberrant myelopoiesis observed in cancer patients, (2) discuss the mechanisms underlying these clinical manifestations and the impact of metabolic perturbations on clinical outcomes, and (3) explore new biomarkers and therapeutic strategies to restore immunometabolism and differentiation of myeloid cells towards an effector phenotype to increase host antitumor immunity. We propose that the profound metabolic alterations and associated transcriptional changes triggered by chronic and overactivated immune responses in myeloid cells represent critical factors influencing the balance between therapeutic efficacy and immune-related adverse effects (irAEs) for current therapeutic strategies, including immune checkpoint inhibitor (ICI) therapy.
Collapse
|
16
|
Zhang T, Fang Z, Linghu KG, Liu J, Gan L, Lin L. Small molecule-driven SIRT3-autophagy-mediated NLRP3 inflammasome inhibition ameliorates inflammatory crosstalk between macrophages and adipocytes. Br J Pharmacol 2020; 177:4645-4665. [PMID: 32726464 DOI: 10.1111/bph.15215] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/03/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE IL-1β produced by macrophages via the NOD-, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome, mediates the inflammatory crosstalk between macrophages and adipocytes. In our previous study, (16S,20S,24R)-12β-acetoxy-16,23-epoxy-24,25-dihydroxy-3β-(β-D-xylopyranosyloxy)-9,19-cyclolanost-22(23)-ene (AEDC), a cycloartane triterpenoid isolated from Actaea vaginata (Ranunculaceae), was found to possess anti-inflammatory effect on LPS-treated RAW264.7 macrophages. This study was designed to investigate whether AEDC modulates macrophage-adipocyte crosstalk to alleviate adipose tissue inflammation. EXPERIMENTAL APPROACH The anti-inflammatory effect of AEDC was evaluated on LPS plus ATP-induced THP-1 macrophages and C57BL/6J mice. The expression of autophagy-related and NLRP3 inflammasome complex proteins was analysed by western blots, immunofluorescence staining and co-immunoprecipitation. The pro-inflammatory cytokines levels were determined by ELISA kits. The adipose tissue inflammation was evaluated by histological analysis and immunohistochemical staining. KEY RESULTS AEDC (5 and 10 μM) activated autophagy, which in turn suppressed the NLRP3 inflammasome activation and IL-1β secretion in THP-1 macrophages. AEDC increased the expression of SIRT3 deacetylase and enhanced its deacetylating activity to reverse mitochondrial dysfunction and activate AMP-activated protein kinase, which together induced autophagy. Moreover, AEDC (10 μM) attenuated macrophage conditioned medium-induced inflammatory responses in adipocytes and blocked THP-1 macrophages migration towards 3T3-L1 adipocytes. In inflammation mice, AEDC (5 and 20 mg·kg-1 ) treatment reduced the levels of pro-inflammatory cytokines in serum and epididymal adipose tissue and reduced macrophage infiltration to alleviate adipose tissue inflammation. CONCLUSION AND IMPLICATIONS AEDC attenuated the inflammatory crosstalk between macrophages and adipocytes through SIRT3-autophagy-mediated NLRP3 inflammasome inhibition, which might used for the treatment of adipose tissue inflammation-related metabolic disorders.
Collapse
Affiliation(s)
- Tian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Zhujun Fang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ke-Gang Linghu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Jingxin Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Lishe Gan
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| |
Collapse
|
17
|
Liu Q, Yu J, Wang L, Tang Y, Zhou Q, Ji S, Wang Y, Santos L, Haeusler RA, Que J, Rajbhandari P, Lei X, Valenti L, Pajvani UB, Qin J, Qiang L. Inhibition of PU.1 ameliorates metabolic dysfunction and non-alcoholic steatohepatitis. J Hepatol 2020; 73:361-370. [PMID: 32135178 DOI: 10.1016/j.jhep.2020.02.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Obesity is a well-established risk factor for type 2 diabetes (T2D) and non-alcoholic steatohepatitis (NASH), but the underlying mechanisms remain incompletely understood. Herein, we aimed to identify novel pathogenic factors (and possible therapeutic targets) underlying metabolic dysfunction in the liver. METHODS We applied a tandem quantitative proteomics strategy to enrich and identify transcription factors (TFs) induced in the obese liver. We used flow cytometry of liver cells to analyze the source of the induced TFs. We employed conditional knockout mice, shRNA, and small-molecule inhibitors to test the metabolic consequences of the induction of identified TFs. Finally, we validated mouse data in patient liver biopsies. RESULTS We identified PU.1/SPI1, the master hematopoietic regulator, as one of the most upregulated TFs in livers from diet-induced obese (DIO) and genetically obese (db/db) mice. Targeting PU.1 in the whole liver, but not hepatocytes alone, significantly improved glucose homeostasis and suppressed liver inflammation. Consistently, treatment with the PU.1 inhibitor DB1976 markedly reduced inflammation and improved glucose homeostasis and dyslipidemia in DIO mice, and strongly suppressed glucose intolerance, liver steatosis, inflammation, and fibrosis in a dietary NASH mouse model. Furthermore, hepatic PU.1 expression was positively correlated with insulin resistance and inflammation in liver biopsies from patients. CONCLUSIONS These data suggest that the elevated hematopoietic factor PU.1 promotes liver metabolic dysfunction, and may be a useful therapeutic target for obesity, insulin resistance/T2D, and NASH. LAY SUMMARY Expression of the immune regulator PU.1 is increased in livers of obese mice and people. Blocking PU.1 improved glucose homeostasis, and reduced liver steatosis, inflammation and fibrosis in mouse models of non-alcoholic steatohepatitis. Inhibition of PU.1 is thus a potential therapeutic strategy for treating obesity-associated liver dysfunction and metabolic diseases.
Collapse
Affiliation(s)
- Qiongming Liu
- Naomi Berrie Diabetes Center, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, 10032, USA; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (The PHOENIX Center at Beijing), Beijing 102206, China
| | - Junjie Yu
- Naomi Berrie Diabetes Center, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, 10032, USA
| | - Liheng Wang
- Naomi Berrie Diabetes Center, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, 10032, USA
| | - Yuliang Tang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Quan Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (The PHOENIX Center at Beijing), Beijing 102206, China
| | - Shuhui Ji
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (The PHOENIX Center at Beijing), Beijing 102206, China
| | - Yi Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Luis Santos
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, 10032, USA
| | - Jianwen Que
- Columbia Center for Human Development and Department of Medicine, Columbia University, New York, NY 10032
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Utpal B Pajvani
- Naomi Berrie Diabetes Center, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, 10032, USA.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (The PHOENIX Center at Beijing), Beijing 102206, China; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030, USA.
| | - Li Qiang
- Naomi Berrie Diabetes Center, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, 10032, USA.
| |
Collapse
|
18
|
Abstract
Spirulina, a cyanobacteria commonly referred to as a blue-green algae, is one of the oldest lifeforms on Earth. Spirulina grows in both fresh and saltwater sources and is known for its high protein and micronutrient content. This review paper will cover the effects of spirulina on weight loss and blood lipids. The currently literature supports the benefits of spirulina for reducing body fat, waist circumference, body mass index and appetite and shows that spirulina has significant benefits for improving blood lipids.
Collapse
Affiliation(s)
| | - Anusha G Bhat
- Department of Internal Medicine, Baystate Medical Center, Springfield, Massachusetts, USA
- Department of Public Heath Practice, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts, United States
| | - James OKeefe
- Saint Lukes Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
19
|
Lackey DE, Reis FCG, Isaac R, Zapata RC, El Ouarrat D, Lee YS, Bandyopadhyay G, Ofrecio JM, Oh DY, Osborn O. Adipocyte PU.1 knockout promotes insulin sensitivity in HFD-fed obese mice. Sci Rep 2019; 9:14779. [PMID: 31611602 PMCID: PMC6791934 DOI: 10.1038/s41598-019-51196-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 09/26/2019] [Indexed: 12/27/2022] Open
Abstract
Insulin resistance is a key feature of obesity and type 2 diabetes. PU.1 is a master transcription factor predominantly expressed in macrophages but after HFD feeding PU.1 expression is also significantly increased in adipocytes. We generated adipocyte specific PU.1 knockout mice using adiponectin cre to investigate the role of PU.1 in adipocyte biology, insulin and glucose homeostasis. In HFD-fed obese mice systemic glucose tolerance and insulin sensitivity were improved in PU.1 AKO mice and clamp studies indicated improvements in both adipose and liver insulin sensitivity. At the level of adipose tissue, macrophage infiltration and inflammation was decreased and glucose uptake was increased in PU.1 AKO mice compared with controls. While PU.1 deletion in adipocytes did not affect the gene expression of PPARg itself, we observed increased expression of PPARg target genes in eWAT from HFD fed PU.1 AKO mice compared with controls. Furthermore, we observed decreased phosphorylation at serine 273 in PU.1 AKO mice compared with fl/fl controls, indicating that PPARg is more active when PU.1 expression is reduced in adipocytes. Therefore, in obesity the increased expression of PU.1 in adipocytes modifies the adipocyte PPARg cistrome resulting in impaired glucose tolerance and insulin sensitivity.
Collapse
Affiliation(s)
- Denise E Lackey
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Felipe C G Reis
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Roi Isaac
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Rizaldy C Zapata
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Dalila El Ouarrat
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Yun Sok Lee
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Gautam Bandyopadhyay
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Jachelle M Ofrecio
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Da Young Oh
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, USA
| | - Olivia Osborn
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Shen S, Liao Q, Zhang T, Pan R, Lin L. Myricanol modulates skeletal muscle-adipose tissue crosstalk to alleviate high-fat diet-induced obesity and insulin resistance. Br J Pharmacol 2019; 176:3983-4001. [PMID: 31339170 DOI: 10.1111/bph.14802] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Skeletal muscle is the predominant site for glucose disposal and fatty acid consumption. Emerging evidence indicates that the crosstalk between adipose tissue and skeletal muscle is critical in maintaining insulin sensitivity and lipid homeostasis. The current study was designed to investigate whether myricanol improves insulin sensitivity and alleviates adiposity through modulating skeletal muscle-adipose tissue crosstalk. EXPERIMENTAL APPROACH The therapeutic effect of myricanol was evaluated on palmitic acid (PA)-treated C2C12 myotubes and high-fat diet (HFD)-fed mice. The crosstalk between myotubes and adipocytes was evaluated using Transwell assay. The cellular lipid content was examined by Nile red staining. The mitochondrial content was assessed by MitoTracker Green staining and citrate synthase activity, and the mitochondrial function was examined by Seahorse assay. Expression of mitochondria-related and insulin signalling pathway proteins was analysed by Western blot, and the irisin level was determined by elisa kit. KEY RESULTS Myricanol increased mitochondrial quantity and function through activating AMP-activated protein kinase, resulting in reduced lipid accumulation and enhanced insulin-stimulated glucose uptake, in PA-treated C2C12 myotubes. Furthermore, myricanol stimulated irisin production and secretion from myotubes to reduce lipid content in 3T3-L1 adipocytes. In HFD-fed mice, myricanol treatment alleviated adiposity and insulin resistance through enhancing lipid utilization and irisin production in skeletal muscle and inducing browning of inguinal fat. CONCLUSIONS AND IMPLICATIONS Myricanol modulates skeletal muscle-adipose tissue crosstalk, to stimulate browning of adipose tissue and improve insulin sensitivity in skeletal muscle. Myricanol might be a potential candidate for treating insulin resistance and obesity.
Collapse
Affiliation(s)
- Shengnan Shen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Qiwen Liao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Tian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Ruile Pan
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
21
|
Sharma NK, Chuang Key CC, Civelek M, Wabitsch M, Comeau ME, Langefeld CD, Parks JS, Das SK. Genetic Regulation of Enoyl-CoA Hydratase Domain-Containing 3 in Adipose Tissue Determines Insulin Sensitivity in African Americans and Europeans. Diabetes 2019; 68:1508-1522. [PMID: 31010960 PMCID: PMC6609988 DOI: 10.2337/db18-1229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/03/2019] [Indexed: 12/17/2022]
Abstract
Insulin resistance (IR) is a harbinger of type 2 diabetes (T2D) and partly determined by genetic factors. However, genetically regulated mechanisms of IR remain poorly understood. Using gene expression, genotype, and insulin sensitivity data from the African American Genetics of Metabolism and Expression (AAGMEx) cohort, we performed transcript-wide correlation and expression quantitative trait loci (eQTL) analyses to identify IR-correlated cis-regulated transcripts (cis-eGenes) in adipose tissue. These IR-correlated cis-eGenes were tested in the European ancestry individuals in the Metabolic Syndrome in Men (METSIM) cohort for trans-ethnic replication. Comparison of Matsuda index-correlated transcripts in AAGMEx with the METSIM study identified significant correlation of 3,849 transcripts, with concordant direction of effect for 97.5% of the transcripts. cis-eQTL for 587 Matsuda index-correlated genes were identified in both cohorts. Enoyl-CoA hydratase domain-containing 3 (ECHDC3) was the top-ranked Matsuda index-correlated cis-eGene. Expression levels of ECHDC3 were positively correlated with Matsuda index, and regulated by cis-eQTL, rs34844369 being the top cis-eSNP in AAGMEx. Silencing of ECHDC3 in adipocytes significantly reduced insulin-stimulated glucose uptake and Akt Ser473 phosphorylation. RNA sequencing analysis identified 691 differentially expressed genes in ECHDC3-knockdown adipocytes, which were enriched in γ-linolenate biosynthesis, and known IR genes. Thus, our studies elucidated genetic regulatory mechanisms of IR and identified genes and pathways in adipose tissue that are mechanistically involved in IR.
Collapse
Affiliation(s)
- Neeraj K Sharma
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Chia-Chi Chuang Key
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Mete Civelek
- Center for Public Health Genomics, Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Mary E Comeau
- Division of Public Health Sciences, Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | - Carl D Langefeld
- Division of Public Health Sciences, Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | - John S Parks
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Swapan K Das
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
22
|
Ronis MJ, Blackburn ML, Shankar K, Ferguson M, Cleves MA, Badger TM. Estradiol and NADPH oxidase crosstalk regulates responses to high fat feeding in female mice. Exp Biol Med (Maywood) 2019; 244:834-845. [PMID: 31161785 DOI: 10.1177/1535370219853563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We previously demonstrated protection against high fat-induced obesity in female but not male p47phox−/− mice lacking NADPH oxidase NOX1/2 activity. To test the role of estradiol (E2)-NOX crosstalk in development of this sexually dimorphic phenotype, we fed diets containing 42% fat/0.5% cholesterol to intact and ovariectomized wild type female C57BL/6 mice and female p47phox−/− mice and to ovariectomized mice where the diet was supplemented with an 1 mg/kg 17β estradiol (E2) for 12 weeks from PND28. Weight gain, gonadal fat pad weight, serum leptin and adiponectin, and adipose tissue inflammation were greater in intact wild type vs. p47 mice ( P < 0.05). Genotype effects on body weight/fat mass were abolished after ovariectomized and restored in OVX + E2 mice ( P < 0.05). The mRNA of downstream PPARγ targets CD36, lipoprotein lipase, and leptin was higher in intact wild type vs. p47phox−/− mice mice ( P < 0.05). Likewise, intact high fat-fed wild type mice had higher expression of the cytokine Mcp1; the pyroptosis marker Nirp3 and matrix remodeling and fibrosis markers Mmp2, Col1A1, and Col6a3 mRNAs ( P < 0.05). These genotype effects were reversed and restored by ovariectomized and OVX + E2, respectively ( P < 0.05). These data suggest that triglyceride accumulation in adipose tissue and development of adipose tissue injury in response to feeding diets high in fat and cholesterol is regulated by the balance between NOX-dependent reactive oxygen species signaling and E2-signaling during development. Loss of estrogens post menopause may increase the risk of obesity and metabolic syndrome as the result disinhibition of reactive oxygen species signaling. Impact statement Estrogens are known to regulate body composition. In addition, reactive oxygen species (ROS) produced by the action of NADPH oxidase (NOX) enzymes have been linked to obesity development. We examined development of obesity and adipose tissue injury in response to feeding “Western” diets high in fat and cholesterol in intact, ovariectomized (OVX), and estrogen-replaced (OVX + E2) wild type and p47phox−/− female mice where NOX2 activity is inhibited. Weight gain, gonadal fat pad weight, and adipose tissue inflammation were greater in intact WT vs. p47phox−/− mice. Genotype effects on body weight/fat mass were abolished after OVX and restored in OVX + E2 mice. These data indicate adipose tissue responses to feeding the “Western” diet is regulated by negative cross-talk between NOX-dependent ROS signaling and E2-signaling during development. Loss of estrogens post menopause may increase the risk of obesity and metabolic syndrome as the result disinhibition of ROS signaling.
Collapse
Affiliation(s)
- Martin J Ronis
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA.,3 Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michael L Blackburn
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Kartik Shankar
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Matthew Ferguson
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Mario A Cleves
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Thomas M Badger
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| |
Collapse
|
23
|
Zhang T, Liu J, Shen S, Tong Q, Ma X, Lin L. SIRT3 promotes lipophagy and chaperon-mediated autophagy to protect hepatocytes against lipotoxicity. Cell Death Differ 2019; 27:329-344. [PMID: 31160717 PMCID: PMC7206074 DOI: 10.1038/s41418-019-0356-z] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
Lipophagy is a lysosomal lipolytic pathway that complements the actions of cytosolic neutral lipases. Chaperon-mediated autophagy (CMA) triggers lipid droplets (LDs) breakdown, to initiate lipolysis via either cytosolic lipases or macroautophagy. SIRT3, a mitochondrial NAD+-dependent deacetylase, regulates the acetylation status and activity of many substrates involving in energy metabolism. However, the role of SIRT3 in regulating lipophagy is controversial. The current study showed that SIRT3 expression was decreased and the macroautophagy flux was blocked in the primary hepatocytes from high-fat diet fed mice and P/O (palmitic acid and oleic acid mixture) treated AML12 mouse hepatocytes, compared with the corresponding controls. SIRT3 overexpression promoted macroautophagy in LDs from P/O-treated hepatocytes through activating AMP-activated protein kinase (AMPK) and unc-51-like kinase 1, to boost LDs digestion. Gain of SIRT3 expression stimulated the formation of lysosome-associated membrane protein 2A (LAMP-2A)-heat shock cognate 71 kDa protein (HSC70)-perilipin-2 (PLN2) complex, to promote CMA process and reduce the stability of LDs in hepatocytes. Moreover, SIRT3 reduced the expression of stearoyl-CoA desaturase 1, to suppress lipogenesis. In addition, SIRT3 overexpression promoted LDs dispersion on detyrosinated microtubules, and directly deacetylated long-chain acyl-CoA dehydrogenase to enhance mitochondrial energetics. Taken together, SIRT3 ameliorates lipotoxicity in hepatocytes, which might be a potential target for the treatment of nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Tian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jingxin Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Shengnan Shen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Qiang Tong
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Xiaojun Ma
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
24
|
DiNicolantonio JJ, McCarty M, OKeefe J. Does elevated bilirubin aid weight control by preventing development of hypothalamic leptin resistance? Open Heart 2019; 6:e000897. [PMID: 30997121 PMCID: PMC6443125 DOI: 10.1136/openhrt-2018-000897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
| | | | - James OKeefe
- Preventive Cardiology, Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
25
|
Activated glycine receptors may decrease endosomal NADPH oxidase activity by opposing ClC-3-mediated efflux of chloride from endosomes. Med Hypotheses 2019; 123:125-129. [PMID: 30696582 DOI: 10.1016/j.mehy.2019.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/15/2019] [Indexed: 12/25/2022]
|
26
|
Elbere I, Silamikelis I, Ustinova M, Kalnina I, Zaharenko L, Peculis R, Konrade I, Ciuculete DM, Zhukovsky C, Gudra D, Radovica-Spalvina I, Fridmanis D, Pirags V, Schiöth HB, Klovins J. Significantly altered peripheral blood cell DNA methylation profile as a result of immediate effect of metformin use in healthy individuals. Clin Epigenetics 2018; 10:156. [PMID: 30545422 PMCID: PMC6293577 DOI: 10.1186/s13148-018-0593-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Metformin is a widely prescribed antihyperglycemic agent that has been also associated with multiple therapeutic effects in various diseases, including several types of malignancies. There is growing evidence regarding the contribution of the epigenetic mechanisms in reaching metformin's therapeutic goals; however, the effect of metformin on human cells in vivo is not comprehensively studied. The aim of our study was to examine metformin-induced alterations of DNA methylation profiles in white blood cells of healthy volunteers, employing a longitudinal study design. RESULTS Twelve healthy metformin-naïve individuals where enrolled in the study. Genome-wide DNA methylation pattern was estimated at baseline, 10 h and 7 days after the start of metformin administration. The whole-genome DNA methylation analysis in total revealed 125 differentially methylated CpGs, of which 11 CpGs and their associated genes with the most consistent changes in the DNA methylation profile were selected: POFUT2, CAMKK1, EML3, KIAA1614, UPF1, MUC4, LOC727982, SIX3, ADAM8, SNORD12B, VPS8, and several differentially methylated regions as novel potential epigenetic targets of metformin. The main functions of the majority of top-ranked differentially methylated loci and their representative cell signaling pathways were linked to the well-known metformin therapy targets: regulatory processes of energy homeostasis, inflammatory responses, tumorigenesis, and neurodegenerative diseases. CONCLUSIONS Here we demonstrate for the first time the immediate effect of short-term metformin administration at therapeutic doses on epigenetic regulation in human white blood cells. These findings suggest the DNA methylation process as one of the mechanisms involved in the action of metformin, thereby revealing novel targets and directions of the molecular mechanisms underlying the various beneficial effects of metformin. TRIAL REGISTRATION EU Clinical Trials Register, 2016-001092-74. Registered 23 March 2017, https://www.clinicaltrialsregister.eu/ctr-search/trial/2016-001092-74/LV .
Collapse
Affiliation(s)
- Ilze Elbere
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ivars Silamikelis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Monta Ustinova
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ineta Kalnina
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Linda Zaharenko
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Raitis Peculis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ilze Konrade
- Riga East Clinical University Hospital, 2 Hipokrata Street, Riga, LV-1038, Latvia
| | - Diana Maria Ciuculete
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Christina Zhukovsky
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Dita Gudra
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ilze Radovica-Spalvina
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Davids Fridmanis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Valdis Pirags
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia.
| |
Collapse
|
27
|
Wang A, Zhou F, Li D, Lu JJ, Wang Y, Lin L. γ-Mangostin alleviates liver fibrosis through Sirtuin 3-superoxide-high mobility group box 1 signaling axis. Toxicol Appl Pharmacol 2018; 363:142-153. [PMID: 30502394 DOI: 10.1016/j.taap.2018.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022]
Abstract
The activation of hepatic stellate cells (HSCs) plays a critical role in liver fibrosis. In the current study, γ-mangostin (γ-man), one of the major xanthones from mangosteen (Garcinia mangostana), was found to alleviate fibrogenesis in human immortalized HSCs (LX-2 cells) and in liver from chronic carbon tetrachloride (CCl4) injured mice. γ-Man suppressed the expression levels of collagen I and α-smooth muscle actin (α-SMA) in LX-2 cells in both dose and time dependent manners. Furthermore, γ-man inhibited NAD(P)H oxidase activity through induction of sirtuin 3 (SIRT3), resulting in reduced intracellular oxidative stress in LX-2 cells. Moreover, γ-man stimulated the expression of histone deacetylase 1, which in turn decreased the acetylation and cytoplasmic shuttling of high mobility group box 1 (HMGB1), to impair autocrine HMGB1-induced HSC activation. In CCl4-injured mice, γ-man enhanced the expression of SIRT3 and decreased the expression of HMGB1, resulting in decreased accumulation of collagen I and α-SMA in liver. Consequently, γ-man might be a potent candidate to treat oxidative stress induced liver fibrosis.
Collapse
Affiliation(s)
- Anqi Wang
- Guangdong-Macau Traditional Chinese Medicine Technology Industrial Park Development Co., Ltd, Hengqin New Area, Zhuhai, Guangdong 519031, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Fayang Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Dan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
28
|
Saxena A, Sachin K. A Network Biology Approach for Assessing the Role of Pathologic Adipose Tissues in Insulin Resistance Using Meta-analysis of Microarray Datasets. Curr Genomics 2018; 19:630-666. [PMID: 30386174 PMCID: PMC6194434 DOI: 10.2174/1389202919666180726125645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 05/10/2018] [Accepted: 07/16/2018] [Indexed: 11/22/2022] Open
Abstract
Background The role of adipose tissue in Insulin resistance (IR) and Type 2 Diabetes (T2D) has well been received in the biomedical community; being a precursor of T2D, identification of the molecular basis of IR is therefore, vital to elucidate T2D- pathogenesis and meta-analysis of previously conducted microarray studies provides an inexpensive approach to achieve this end. Methods In this study, we have carried out a statistical meta-analysis of 157 microarray datasets from five independent studies and identified a meta-signature of 1,511 genes; their functional meaning was elucidated by integrated pathways-analysis. Further, a protein-protein interaction network was constructed and key genes along with their high confidence transcriptional- and epigenetic-mediators were identified using a network biology approach. Results Various inflammation- and immune system-related pathways such as TGF-β signaling, IL7 signaling, Neutrophil degranulation, and Chemokine signaling etc. were enriched in sick adipose tissues; identified transcription factors, and microRNAs were also found to regulate processes relevant to IR/T2D pathophysiology. Conclusion This study endorses the development of effective bioinformatics workflow and further grants an indication for the acceptance of adiposopathy as the root mechanistic pathology that poses risk for development of type 2 diabetes; concept of adipospathy in place of metabolic syndrome will open the possibility to design drugs, those will ameliorate adipose functions and hence proved to be more effective against Type 2 Diabetes.
Collapse
Affiliation(s)
- Aditya Saxena
- 1Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura (U.P.), India; 2Uttarakhand Technical University, Dehradun (U.K.), India; 3Department of Biochemistry and Biotechnology, S.B.S. (PG) Institute of Biomedical Sciences & Research, Dehradun (U.K.), India
| | - Kumar Sachin
- 1Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura (U.P.), India; 2Uttarakhand Technical University, Dehradun (U.K.), India; 3Department of Biochemistry and Biotechnology, S.B.S. (PG) Institute of Biomedical Sciences & Research, Dehradun (U.K.), India
| |
Collapse
|
29
|
DiNicolantonio JJ, McCarty MF, O’Keefe JH. Antioxidant bilirubin works in multiple ways to reduce risk for obesity and its health complications. Open Heart 2018; 5:e000914. [PMID: 30364545 PMCID: PMC6196942 DOI: 10.1136/openhrt-2018-000914] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- James J DiNicolantonio
- Department of Preventive Cardiology, Saint Luke’s Mid America Heart Institute, Kansas City, Missouri, USA
| | | | - James H O’Keefe
- Department of Preventive Cardiology, Saint Luke’s Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
30
|
aP2-Cre Mediated Ablation of GHS-R Attenuates Adiposity and Improves Insulin Sensitivity during Aging. Int J Mol Sci 2018; 19:ijms19103002. [PMID: 30275401 PMCID: PMC6213105 DOI: 10.3390/ijms19103002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 01/08/2023] Open
Abstract
Ghrelin via its receptor, the growth hormone secretagogue receptor (GHS-R), increases food intake and adiposity. The tissue-specific functions of GHS-R in peripheral tissues are mostly unknown. We previously reported that while GHS-R expression is very low in white and brown fat of young mice, expression increases during aging. To investigate whether GHS-R has cell-autonomous effects in adipose tissues, we generated aP2-Cre-mediated GHS-R knockdown mice (aP2-Cre/Ghsrf/f). We studied young (5–6 months) and old (15–17 months) aP2-Cre/Ghsrf/f mice and their age-matched controls. Interestingly, young aP2-Cre/Ghsrf/f mice had normal body weight but reduced fat; old mice showed pronounced reductions of both body weight and body fat. Calorimetry analysis revealed that aP2-Cre/Ghsrf/f mice had normal food intake and locomotor activity at both young and old age; but intriguingly, while energy expenditure was normal at young age, it was significantly increased at old age. Both young and old aP2-Cre/Ghsrf/f mice exhibited improved insulin sensitivity and glucose tolerance. Importantly, old aP2-Cre/Ghsrf/f mice maintained higher core body temperature at 4 °C, and showed higher expression of the thermogenic uncoupling protein 1 (UCP1) gene. The ex vivo studies further demonstrated that GHS-R deficient white adipocytes from old mice exhibit increased glucose uptake and lipolysis, promoting lipid mobilization. Despite the fact that the in vivo phenotypes of aP2-Cre/Ghsrf/f mice may not be exclusively determined by GHS-R knockdown in adipose tissues, our data support that GHS-R has cell-autonomous effects in adipocytes. The anabolic effect of GHS-R in adipocytes is more pronounced in aging, which likely contributes to age-associated obesity and insulin resistance.
Collapse
|
31
|
Abstract
Diabetes is a chronic metabolic disorder that poses a global burden to healthcare. Increasing incidence of diabetes-related complications in the affected population includes a delay in wound healing that often results in non-traumatic limb amputations. Owing to the intricacies of the healing process and crosstalk between the multitude of participating cells, the identification of hyperglycaemia-induced changes at both cellular and molecular levels poses a challenge. Macrophages are one of the key participants in wound healing and continue to exert functional changes at the wound site since the time of injury. In the present review, we discuss the role of these cells and their aberrant functions in diabetic wounds. We have extensively studied the process of macrophage polarization (MP) and its modulation through epigenetic modifications. Data from both pre-clinical and clinical studies on diabetes have co-related hyperglycaemia induced changes in gene expression to an increased incidence of diabetic complications. Hyperglycaemia and oxidative stress, create an environment prone to changes in the epigenetic code, that is manifested as an altered inflammatory gene expression. Here, we have attempted to understand the different epigenetic modulations that possibly contribute towards dysregulated MP, resulting in delayed wound healing.
Collapse
Affiliation(s)
- Sanchari Basu Mallik
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (formerly Manipal University), Manipal 576104, Karnataka, India
| | - B S Jayashree
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (formerly Manipal University), Manipal 576104, Karnataka, India
| | - Rekha R Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (formerly Manipal University), Manipal 576104, Karnataka, India.
| |
Collapse
|
32
|
Li D, Liu Q, Sun W, Chen X, Wang Y, Sun Y, Lin L. 1,3,6,7-Tetrahydroxy-8-prenylxanthone ameliorates inflammatory responses resulting from the paracrine interaction of adipocytes and macrophages. Br J Pharmacol 2018; 175:1590-1606. [PMID: 29446826 DOI: 10.1111/bph.14162] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/29/2017] [Accepted: 01/18/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic inflammation in adipose tissue is critical in the onset and development of insulin resistance and type 2 diabetes. Macrophage infiltration into adipose tissue and pro-inflammatory polarization play key roles in adipose tissue inflammation. The fruit hull of mangosteen (Garcinia mangostana) is used in traditional medicine to treat various inflammatory diseases. However, its role in regulating adipose tissue inflammation is unexplored. This study was designed to identify xanthones from G. mangostana, which could ameliorate adipose tissue inflammation. EXPERIMENTAL APPROACH Expressions of inducible NOS, cytokines, chemokines and components of the NF-κB and MAPKs pathways were evaluated using Western blotting, immunofluorescence, quantitative real-time PCR or ELISA. The migration of macrophages towards adipocytes was tested using Transwell experiments in vitro. A murine model of LPS-induced acute inflammation was used to examine effects of 1,3,6,7-tetrahydroxy-8-prenylxanthone (TPX) on inflammatory responses in adipose tissue in vivo. KEY RESULTS From a series of xanthones isolated from G. mangostana, TPX was identified as a potent inhibitor of LPS-induced NO production and IL-6 secretion in RAW264.7 macrophages. TPX ameliorated LPS-induced inflammatory responses in RAW264.7 macrophages, and TNF-α-mediated inflammation in 3T3-L1 adipocytes, through inhibiting MAPKs and NF-κB activation and promoting sirtuin 3 expression. TPX also blocked RAW264.7 macrophages migration towards 3T3-L1 adipocytes in co-cultures. Furthermore, TPX alleviated LPS-induced adipose tissue inflammation in vivo by reducing pro-inflammatory cytokines and preventing the pro-inflammatory polarization of macrophages. CONCLUSIONS AND IMPLICATIONS Taken together, our results indicate that TPX disrupts the inflammatory responses between macrophages and adipocytes, and attenuates adipose tissue inflammation.
Collapse
Affiliation(s)
- Dan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Qianyu Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Wen Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
33
|
Liu J, Li D, Zhang T, Tong Q, Ye RD, Lin L. SIRT3 protects hepatocytes from oxidative injury by enhancing ROS scavenging and mitochondrial integrity. Cell Death Dis 2017; 8:e3158. [PMID: 29072685 PMCID: PMC5680927 DOI: 10.1038/cddis.2017.564] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/17/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022]
Abstract
Evidences of oxidative stress and mitochondrial dysfunction have been recognized in most of clinical and experimental liver diseases. SIRT3, a member of NAD+-dependent deacetylases, is mainly localized in mitochondria. So far, the role of SIRT3 in protecting hepatocytes against oxidative stress remains elusive. Herein, we found SIRT3 protein expression is decreased in tert-butyl hydroperoxide (t-BHP)-treated AML12 cells in vitro and primary hepatocytes from CCl4-injured mice in vivo. To further verify the role of SIRT3 in protecting hepatocytes from t-BHP-induced injury, SIRT3 overexpressed AML12 cell line and primary hepatocytes were generated. SIRT3 overexpressed hepatocytes showed improved cell viability upon t-BHP challenge, with less intracellular reactive oxygen species (ROS) accumulation. SIRT3 overexpression reduced superoxide dismutase 2 acetylation level and stimulated nuclear factor erythroid 2-related factor 2 nuclear translocation to enhance anti-oxidative capacity. Moreover, SIRT3 deacetylated peroxisome proliferator-activated receptor γ coactivator 1α to promote mitochondrial biogenesis, and 8-oxoguanine DNA glycosylase 1 to orchestrate DNA repair, resulting in improved mitochondrial function. Through deacetylating Ku70, SIRT3 also abated mitochondrial translocation of dynamin-related protein 1, to attenuate mitochondrial fragmentation in t-BHP-injured hepatocytes. These results suggested that SIRT3 protected hepatocytes against oxidative stress by enhancing ROS scavenging and maintaining mitochondrial integrity.
Collapse
Affiliation(s)
- Jingxin Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Dan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Tian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Richard Dequan Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau, China
| |
Collapse
|
34
|
Bjørklund G, Chirumbolo S. Role of oxidative stress and antioxidants in daily nutrition and human health. Nutrition 2016; 33:311-321. [PMID: 27746034 DOI: 10.1016/j.nut.2016.07.018] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/14/2016] [Accepted: 07/24/2016] [Indexed: 12/20/2022]
Abstract
Diet may be defined as a complex process that should involve a deeper comprehension of metabolism, energy balance, and the molecular pathways involved in cellular stress response and survival, gut microflora genetics, enzymatic polymorphism within the human population, and the role of plant-derived polyphenols in this context. Metabolic syndrome, encompassing pathologies with a relatively high morbidity, such as type 2 diabetes, obesity, and cardiovascular disease, is a bullet point of the big concern about how daily dietary habits should promote health and prevent metabolic impairments to prevent hospitalization and the need for health care. From a clinical point of view, very few papers deal with this concern, whereas most of the evidence reported focuses on in vitro and animal models, which study the activity of phytochemicals contained in the daily diet. A fundamental issue addressed by dietitians deals with the role exerted by redox-derived reactive species. Most plant polyphenols act as antioxidants, but recent evidence supports the idea that these compounds primarily activate a mild oxidative stress to elicit a positive, beneficial response from cells. How these compounds may act upon the detoxifying system exerting a scavenging role from reactive oxygen or nitrogen species is still a matter of debate; however, it can be argued that their role is even more complex than expected, acting as signaling molecules in the cross-talk mitochondria-endoplasmic reticulum and in enzymatic pathways involved in the energetic balance. In this relationship, a fundamental role is played by the brain-adipose tissue-gut axis. The aim of this review was to elucidate this topic and the state of art about the role of reactive species in cell signaling and the function of metabolism and survival to reappraise the role of plant-derived chemicals.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
35
|
Xu Q, Shang Y, Li Y, Zhang F, Gao S, Yao M. MicroRNAs 103 and 107 link type 2 diabetes and post-menopausal breast cancer. Int J Diabetes Dev Ctries 2015. [DOI: 10.1007/s13410-015-0412-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
36
|
Wu Y, Song P, Zhang W, Liu J, Dai X, Liu Z, Lu Q, Ouyang C, Xie Z, Zhao Z, Zhuo X, Viollet B, Foretz M, Wu J, Yuan Z, Zou MH. Activation of AMPKα2 in adipocytes is essential for nicotine-induced insulin resistance in vivo. Nat Med 2015; 21:373-82. [PMID: 25799226 PMCID: PMC4390501 DOI: 10.1038/nm.3826] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/17/2015] [Indexed: 12/15/2022]
Abstract
Cigarette smoking promotes body weight reduction in humans while paradoxically also promoting insulin resistance (IR) and hyperinsulinemia. However, the mechanisms behind these effects are unclear. Here we show that nicotine, a major constituent of cigarette smoke, selectively activates AMP-activated protein kinase α2 (AMPKα2) in adipocytes, which in turn phosphorylates MAP kinase phosphatase-1 (MKP1) at serine 334, initiating its proteasome-dependent degradation. The nicotine-dependent reduction of MKP1 induces the aberrant activation of both p38 mitogen-activated protein kinase and c-Jun N-terminal kinase, leading to increased phosphorylation of insulin receptor substrate 1 (IRS1) at serine 307. Phosphorylation of IRS1 leads to its degradation, protein kinase B inhibition, and the loss of insulin-mediated inhibition of lipolysis. Consequently, nicotine increases lipolysis, which results in body weight reduction, but this increase also elevates the levels of circulating free fatty acids and thus causes IR in insulin-sensitive tissues. These results establish AMPKα2 as an essential mediator of nicotine-induced whole-body IR in spite of reductions in adiposity.
Collapse
Affiliation(s)
- Yue Wu
- 1] Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. [2] Department of Cardiology, Cardiovascular Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ping Song
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Wencheng Zhang
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Junhui Liu
- Department of Cardiology, Cardiovascular Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaoyan Dai
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Zhaoyu Liu
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Qiulun Lu
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Changhan Ouyang
- 1] Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. [2] Key Laboratory of Hubei Province on Cardio-Cerebral Diseases, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Zhonglin Xie
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Zhengxing Zhao
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xiaozhen Zhuo
- Department of Cardiology, Cardiovascular Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Benoit Viollet
- 1] INSERM, U1016, Institut Cochin, Paris, France. [2] CNRS, UMR 8104, Paris, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marc Foretz
- 1] INSERM, U1016, Institut Cochin, Paris, France. [2] CNRS, UMR 8104, Paris, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jiliang Wu
- Key Laboratory of Hubei Province on Cardio-Cerebral Diseases, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Zuyi Yuan
- Department of Cardiology, Cardiovascular Research Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ming-Hui Zou
- 1] Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. [2] Key Laboratory of Hubei Province on Cardio-Cerebral Diseases, Hubei University of Science and Technology, Xianning, Hubei, China
| |
Collapse
|
37
|
Wei N, Wang Y, Xu RX, Wang GQ, Xiong Y, Yu TY, Yang GS, Pang WJ. PU.1antisense lncRNA against its mRNA translation promotes adipogenesis in porcine preadipocytes. Anim Genet 2015; 46:133-40. [DOI: 10.1111/age.12275] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2014] [Indexed: 01/31/2023]
Affiliation(s)
- N. Wei
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Y. Wang
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - R.-X. Xu
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - G.-Q. Wang
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Y. Xiong
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - T.-Y. Yu
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - G.-S. Yang
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| | - W.-J. Pang
- Laboratory of Animal Fat Deposition & Muscle Development; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi 712100 China
| |
Collapse
|
38
|
Lane JM, Doyle JR, Fortin JP, Kopin AS, Ordovás JM. Development of an OP9 derived cell line as a robust model to rapidly study adipocyte differentiation. PLoS One 2014; 9:e112123. [PMID: 25409310 PMCID: PMC4237323 DOI: 10.1371/journal.pone.0112123] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/08/2014] [Indexed: 02/06/2023] Open
Abstract
One hallmark of obesity is adipocyte hypertrophy and hyperplasia. To gain novel insights into adipose biology and therapeutics, there is a pressing need for a robust, rapid, and informative cell model of adipocyte differentiation for potential RNAi and drug screens. Current models are prohibitive for drug and RNAi screens due to a slow differentiation time course and resistance to transfection. We asked if we could create a rapid, robust model of adipogenesis to potentially enable rapid functional and obesity therapeutic screens. We generated the clonal population OP9-K, which differentiates rapidly and reproducibly, and displays classic adipocyte morphology: rounded cell shape, lipid accumulation, and coalescence of lipids into a large droplet. We further validate the OP9-K cells as an adipocyte model system by microarray analysis of the differentiating transcriptome. OP9-K differentiates via known adipogenic pathways, involving the transcriptional activation and repression of common adipose markers Plin1, Gata2, C/Ebpα and C/Ebpβ and biological pathways, such as lipid metabolism, PPARγ signaling, and osteogenesis. We implemented a method to quantify lipid accumulation using automated microscopy and tested the ability of our model to detect alterations in lipid accumulation by reducing levels of the known master adipogenic regulator Pparγ. We further utilized our model to query the effects of a novel obesity therapeutic target, the transcription factor SPI1. We determine that reduction in levels of Spi1 leads to an increase in lipid accumulation. We demonstrate rapid, robust differentiation and efficient transfectability of the OP9-K cell model of adipogenesis. Together with our microscopy based lipid accumulation assay, adipogenesis assays can be achieved in just four days' time. The results of this study can contribute to the development of rapid screens with the potential to deepen our understanding of adipose biology and efficiently test obesity therapeutics.
Collapse
Affiliation(s)
- Jacqueline M. Lane
- Massachusetts General Hospital, Center for Human Genetics Research, 185 Cambridge Street, Boston, MA 02114, United States of America
- Division of Sleep Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, United States of America
- Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, United States of America
- * E-mail:
| | - Jamie R. Doyle
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, 800 Washington St., Box 7703, Boston, MA 02111, United States of America
| | - Jean-Philippe Fortin
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, 800 Washington St., Box 7703, Boston, MA 02111, United States of America
| | - Alan S. Kopin
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, 800 Washington St., Box 7703, Boston, MA 02111, United States of America
| | - José M. Ordovás
- Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, United States of America
| |
Collapse
|
39
|
Vieira-Potter VJ. Inflammation and macrophage modulation in adipose tissues. Cell Microbiol 2014; 16:1484-92. [PMID: 25073615 DOI: 10.1111/cmi.12336] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/24/2014] [Accepted: 07/28/2014] [Indexed: 12/11/2022]
Abstract
The adipose tissue is an active endocrine organ that harbours not only mature and developing adipocytes but also a wide array of immune cells, including macrophages, a key immune cell in determining metabolic functionality. With adipose tissue expansion, M1 pro-inflammatory macrophage infiltration increases, activates other immune cells, and affects lipid trafficking and metabolism, in part via inhibiting mitochondrial function and increasing reactive oxygen species (ROS). The pro-inflammatory cytokines produced and released interfere with insulin signalling, while inhibiting M1 macrophage activation improves systemic insulin sensitivity. In healthy adipose tissue, M2 alternative macrophages predominate and associate with enhanced lipid handling and mitochondrial function, anti-inflammatory cytokine production, and inhibition of ROS. The sequence of events leading to macrophage infiltration and activation in adipose tissue remains incompletely understood but lipid handling of both macrophages and adipocytes appears to play a major role.
Collapse
Affiliation(s)
- Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
40
|
Pang WJ, Lin LG, Xiong Y, Wei N, Wang Y, Shen QW, Yang GS. Knockdown of PU.1 AS lncRNA inhibits adipogenesis through enhancing PU.1 mRNA translation. J Cell Biochem 2013; 114:2500-12. [PMID: 23749759 DOI: 10.1002/jcb.24595] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 05/14/2013] [Indexed: 12/30/2022]
Abstract
PU.1 is an Ets family transcription factor involved in the myelo-lymphoid differentiation. We have previously demonstrated that PU.1 is also expressed in the adipocyte lineage. However, the expression levels of PU.1 mRNA and protein in preadipocytes do not match the levels in mature adipocytes. PU.1 mRNA level is higher in preadipocytes, whereas its protein is expressed in the adipocytes but not in the preadipocytes. The underlying mechanism remains elusive. Here, we find that miR-155 knockdown or overexpression has no effect on the levels of PU.1 mRNA and protein in preadipocytes or adipocytes. MiR-155 regulates adipogenesis not through PU.1, but via C/EBPβ which is another target of miR-155. We also checked the expression levels of PU.1 mRNA and antisense long non-coding RNA (AS lncRNA). Interestingly, compared with the level of PU.1 mRNA, the level of PU.1 AS lncRNA is much higher in preadipocytes, whereas it is opposite in the adipocytes. We further discover that PU.1 AS lncRNA binds to its mRNA forming an mRNA/AS lncRNA compound. The knockdown of PU.1 AS by siRNA inhibits adipogenesis and promotes PU.1 protein expression in both preadipocytes and adipocytes. Furthermore, the repression of PU.1 AS decreases the expression and secretion of adiponectin. We also find that the effect of retroviral-mediated PU.1 AS knockdown on adipogenesis is consistent with that of PU.1 AS knockdown by siRNA. Taken together, our results suggest that PU.1 AS lncRNA promotes adipogenesis through preventing PU.1 mRNA translation via binding to PU.1 mRNA to form mRNA/AS lncRNA duplex in preadipocytes.
Collapse
Affiliation(s)
- Wei-Jun Pang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, 77030
| | | | | | | | | | | | | |
Collapse
|
41
|
Pruning of the adipocyte peroxisome proliferator-activated receptor γ cistrome by hematopoietic master regulator PU.1. Mol Cell Biol 2013; 33:3354-64. [PMID: 23775123 DOI: 10.1128/mcb.00599-13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
"Master" transcription factors are the gatekeepers of lineage identity. As such, they have been a major focus of efforts to manipulate cell fate for therapeutic purposes. The ETS transcription factor PU.1 has a potent ability to confer macrophage phenotypes on cells already committed to a different lineage, but how it overcomes the presence of other master regulators is not known. The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is the master regulator of the adipose lineage, and its genomic binding pattern in adipocytes is well characterized. Here we show that, when expressed at macrophage levels in mature adipocytes, PU.1 bound a large fraction of its macrophage sites, where it induced chromatin opening and the expression of macrophage target genes. Strikingly, PU.1 markedly reduced the genomic binding of PPARγ without changing its abundance. PU.1 expression repressed genes with nearby adipocyte-specific PPARγ binding sites, while a common macrophage-adipocyte gene expression program was retained. Together, these data reveal unexpected lability within the adipocyte PPARγ cistrome and show that, even in terminally differentiated cells, PU.1 can remodel the cistrome of another master regulator.
Collapse
|
42
|
Ronis MJJ, Sharma N, Vantrease J, Borengasser SJ, Ferguson M, Mercer KE, Cleves MA, Gomez-Acevedo H, Badger TM. Female mice lacking p47phox have altered adipose tissue gene expression and are protected against high fat-induced obesity. Physiol Genomics 2013; 45:351-66. [DOI: 10.1152/physiolgenomics.00148.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The current study was designed to determine if the NADPH-oxidase NOX2 plays a role in development of obesity after high fat feeding. Wild-type (WT) mice and mice lacking the essential cytosolic NOX2 system component p47phox (P47KO mice) were fed AIN-93G diets or high-fat diets (HFD) containing 45% fat and 0.5% cholesterol for 13 wk from weaning. Fat mass was increased to a similar degree by HFD in males of both genotypes ( P < 0.05). However, female P47KO-HFD mice had no increase in adiposity or adipocyte size relative to female WT-HFD mice. Resistance to HFD-driven obesity in P47KO females was associated with increased expression of hepatic TFAM and UCP-2 mRNA, markers of mitochondrial number and uncoupling, and increased expression of hepatic mitochondrial respiratory complexes and whole body energy expenditure in response to HFD. Microarray analysis revealed significantly lower expression of mRNA encoding genes linked to energy metabolism, adipocyte differentiation (PPARγ), and fatty acid uptake (CD36, lipoprotein lipase), in fat pads from female P47KO-HFD mice compared with WT-HFD females. Moreover, differentiation of preadipocytes ex vivo was suppressed more by 17β-estradiol in cells from P47KO compared with cells from WT females in conjunction with overexpression of mRNA for Pref-1 ( P < 0.05). HFD mice of both sexes were resistant to the development of hyperglycemia and hepatic steatosis ( P < 0.05) and had reduced serum triglycerides, leptin, and adiponectin relative to WT-HFD mice ( P < 0.05). These data suggest that NOX2 is an important regulator of metabolic homeostasis and diet-induced obesity.
Collapse
Affiliation(s)
- Martin J. J. Ronis
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and
| | - Neha Sharma
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jamie Vantrease
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sarah J. Borengasser
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and
| | - Matthew Ferguson
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kelly E. Mercer
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Mario A. Cleves
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Horacio Gomez-Acevedo
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and
| | - Thomas M. Badger
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
43
|
Wada T, Sunaga H, Ohkawara R, Shimba S. Aryl hydrocarbon receptor modulates NADPH oxidase activity via direct transcriptional regulation of p40phox expression. Mol Pharmacol 2013; 83:1133-40. [PMID: 23478803 DOI: 10.1124/mol.112.083303] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A member of the NADPH oxidase subunits, p40(phox) plays an important role in the regulation of NADPH oxidase activity and the subsequent production of reactive oxygen species (ROS). In this study, we show that mouse p40(phox) is a novel transcriptional target of the aryl hydrocarbon receptor (AhR), known as a dioxin receptor or xenobiotic receptor, in the liver. Treatment of mice with 3-methylcholanthrene (3MC) increased p40(phox) gene expression in the liver, but this induction of p40(phox) gene expression was diminished by the deletion of the AhR gene in the liver. Consistent with the in vivo results, the expression of the p40(phox) gene was increased in 3MC-treated Hepa1c1c7 cells in an AhR-dependent manner. In addition, promoter analysis established p40(phox) as a transcriptional target of AhR. Studies using the RNA-interference technique revealed that p40(phox) is involved in the increase of NADPH oxidase activity and the subsequent ROS production in AhR-activated Hepa1c1c7 cells. Consequently, the results obtained here may provide a novel molecular mechanism for ROS production after exposure to dioxins.
Collapse
Affiliation(s)
- Taira Wada
- Department of Health Science, School of Pharmacy, Nihon University, Funabashi, Chiba, Japan
| | | | | | | |
Collapse
|