42 results on '"Mengwei Zang"'
Search Results
2. Targeting hepatic serine-arginine protein kinase 2 ameliorates alcohol-associated liver disease by alternative splicing control of lipogenesis
- Author
-
Guannan Li, Hanqing Chen, Feng Shen, Steven Blake Smithson, Gavyn Lee Shealy, Qinggong Ping, Zerong Liang, Jingyan Han, Andrew C. Adams, Yu Li, Dechun Feng, Bin Gao, Masahiro Morita, Xianlin Han, Tim H. Huang, Nicolas Musi, and Mengwei Zang
- Subjects
Hepatology - Published
- 2023
3. Age‐dependent loss of hepatic <scp>SIRT1</scp> enhances NLRP3 inflammasome signaling and impairs capacity for liver fibrosis resolution
- Author
-
Jennifer Adjei‐Mosi, Qing Sun, Steven Blake Smithson, Gavyn Lee Shealy, Krupa Dhruvitha Amerineni, Zerong Liang, Hanqing Chen, Mei Wang, Qinggong Ping, Jingyan Han, Masahiro Morita, Amrita Kamat, Nicolas Musi, and Mengwei Zang
- Subjects
Aging ,Cell Biology - Published
- 2023
4. Distinct histopathological phenotypes of severe alcoholic hepatitis suggest different mechanisms driving liver injury and failure
- Author
-
Jing Ma, Adrien Guillot, Zhihong Yang, Bryan Mackowiak, Seonghwan Hwang, Ogyi Park, Brandon J. Peiffer, Ali Reza Ahmadi, Luma Melo, Praveen Kusumanchi, Nazmul Huda, Romil Saxena, Yong He, Yukun Guan, Dechun Feng, Pau Sancho-Bru, Mengwei Zang, Andrew MacGregor Cameron, Ramon Bataller, Frank Tacke, Zhaoli Sun, Suthat Liangpunsakul, and Bin Gao
- Subjects
Inflammation ,Mice, Inbred C57BL ,Mice ,Phenotype ,Ethanol ,Liver ,Hepatitis, Alcoholic ,Animals ,General Medicine ,Reactive Oxygen Species ,Liver Diseases, Alcoholic - Abstract
Intrahepatic neutrophil infiltration has been implicated in severe alcoholic hepatitis (SAH) pathogenesis; however, the mechanism underlying neutrophil-induced injury in SAH remains obscure. This translational study aims to describe the patterns of intrahepatic neutrophil infiltration and its involvement in SAH pathogenesis. Immunohistochemistry analyses of explanted livers identified two SAH phenotypes despite a similar clinical presentation, one with high intrahepatic neutrophils (Neuhi), but low levels of CD8+ T cells, and vice versa. RNA-Seq analyses demonstrated that neutrophil cytosolic factor 1 (NCF1), a key factor in controlling neutrophilic ROS production, was upregulated and correlated with hepatic inflammation and disease progression. To study specifically the mechanisms related to Neuhi in AH patients and liver injury, we used the mouse model of chronic-plus-binge ethanol feeding and found that myeloid-specific deletion of the Ncf1 gene abolished ethanol-induced hepatic inflammation and steatosis. RNA-Seq analysis and the data from experimental models revealed that neutrophilic NCF1-dependent ROS promoted alcoholic hepatitis (AH) by inhibiting AMP-activated protein kinase (a key regulator of lipid metabolism) and microRNA-223 (a key antiinflammatory and antifibrotic microRNA). In conclusion, two distinct histopathological phenotypes based on liver immune phenotyping are observed in SAH patients, suggesting a separate mechanism driving liver injury and/or failure in these patients.
- Published
- 2021
5. LRG1 is an adipokine that mediates obesity-induced hepatosteatosis and insulin resistance
- Author
-
Wenjuan Xia, Fang Zhang, Ying Lv, Mengwei Zang, Feng Dong, Jiyoon Ryu, Lily Q. Dong, Jane L. Lynch, Feng Liu, Paul R. Langlais, Bruce J. Nicholson, Hairong Luo, Jie Wen, Yuguang Shi, Ravindranath Duggirala, Juanhong Liu, Zhe Sun, Sijia He, and Juli Bai
- Subjects
medicine.medical_specialty ,Adipokine ,Mice ,Insulin resistance ,Adipokines ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Humans ,Obesity ,Glycoproteins ,chemistry.chemical_classification ,Mice, Knockout ,biology ,Chemistry ,Fatty Acids ,Fatty acid ,General Medicine ,Metabolism ,medicine.disease ,Fatty Liver ,Insulin receptor ,Endocrinology ,Lipogenesis ,biology.protein ,Insulin Resistance ,Oxidation-Reduction ,Research Article - Abstract
Dysregulation in adipokine biosynthesis and function contributes to obesity-induced metabolic diseases. However, the identities and functions of many of the obesity-induced secretory molecules remain unknown. Here, we report the identification of leucine-rich alpha-2-glycoprotein 1 (LRG1) as an obesity-associated adipokine that exacerbates high fat diet-induced hepatosteatosis and insulin resistance. Serum levels of LRG1 were markedly elevated in obese humans and mice compared with their respective controls. LRG1 deficiency in mice greatly alleviated diet-induced hepatosteatosis, obesity, and insulin resistance. Mechanistically, LRG1 bound with high selectivity to the liver and promoted hepatosteatosis by increasing de novo lipogenesis and suppressing fatty acid β-oxidation. LRG1 also inhibited hepatic insulin signaling by downregulating insulin receptor substrates 1 and 2. Our study identified LRG1 as a key molecule that mediates the crosstalk between adipocytes and hepatocytes in diet-induced hepatosteatosis and insulin resistance. Suppressing LRG1 expression and function may be a promising strategy for the treatment of obesity-related metabolic diseases.
- Published
- 2021
6. Adipose group 1 innate lymphoid cells promote adipose tissue fibrosis and diabetes in obesity
- Author
-
Pengzi Zhang, Xitai Sun, Yan Bi, Xuehui Chu, Lei Shen, Xiao Ye, Mengwei Zang, Hongdong Wang, Fangcen Liu, Can Jiang, Dalong Zhu, Chunping Jiang, Yan Wang, and Wenhuan Feng
- Subjects
0301 basic medicine ,Adoptive cell transfer ,General Physics and Astronomy ,Adipose tissue ,Bariatric Surgery ,02 engineering and technology ,DNA-Activated Protein Kinase ,Fibrosis ,Adipocytes ,Medicine ,Macrophage ,Lymphocytes ,lcsh:Science ,Mice, Knockout ,education.field_of_study ,Multidisciplinary ,Innate lymphoid cell ,Diabetes ,021001 nanoscience & nanotechnology ,DNA-Binding Proteins ,Adipose Tissue ,0210 nano-technology ,Interleukin Receptor Common gamma Subunit ,medicine.medical_specialty ,Science ,Population ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Interferon-gamma ,Immune system ,Internal medicine ,Animals ,Humans ,Obesity ,education ,Innate immune system ,business.industry ,General Chemistry ,Macrophage Activation ,medicine.disease ,Immunity, Innate ,Mice, Inbred C57BL ,Innate immune cells ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,lcsh:Q ,business - Abstract
Pathogenic factors driving obesity to type 2 diabetes (T2D) are not fully understood. Group 1 innate lymphoid cells (ILC1s) are effectors of innate immunity and enriched in inflamed tissues. Here we show that the number of adipose ILC1s increases in obese T2D patients and correlates with glycemic parameters and with the number of ILC1s in the blood; circulating ILC1 numbers decrease as a result of metabolic improvements after bariatric surgery. In vitro co-culture experiments show that human adipose ILC1s promote adipose fibrogenesis and CD11c+ macrophage activation. Reconstruction of the adipose ILC1 population in Prkdc−/−IL2rg−/− mice by adoptive transfer drives adipose fibrogenesis through activation of TGFβ1 signaling; however, transfer of Ifng−/− ILC1s has no effect on adipose fibrogenesis. Furthermore, inhibiting adipose accumulation of ILC1s using IL-12 neutralizing antibodies attenuates adipose tissue fibrosis and improves glycemic tolerance. Our data present insights into the mechanisms of local immune disturbances in obesity-related T2D., Whether adipose ILC1s regulate adipose tissue fibrogenesis is unknown. Here, the authors report a direct role of adipose ILC1s in adipose tissue fibrogenesis and provide insights into the mechanisms of local immune dysfunction in obesity-associated metabolic disorders.
- Published
- 2019
7. Hepatic posttranscriptional network comprised of CCR4–NOT deadenylase and FGF21 maintains systemic metabolic homeostasis
- Author
-
Akinori Takahashi, Sakie Katsumura, René St-Arnaud, Ola Larsson, Vincent Giguère, Tadashi Yamamoto, Hiroshi Kiyonari, Masahiro Morita, Takeshi Nagashima, Nahum Sonenberg, Nadeem Siddiqui, Christopher Rouya, Bahareh Hekmatnejad, Mengwei Zang, Mariko Okada-Hatakeyama, Yuichi Oike, and Ivan Topisirovic
- Subjects
FGF21 ,Protein subunit ,Tristetraprolin ,Regulator ,Adipokine ,Adipose tissue ,Mice, Transgenic ,Biology ,Diet, High-Fat ,03 medical and health sciences ,Paracrine signalling ,Mice ,0302 clinical medicine ,Ribonucleases ,Animals ,Homeostasis ,Humans ,RNA, Messenger ,Cells, Cultured ,030304 developmental biology ,Metabolic Syndrome ,0303 health sciences ,Multidisciplinary ,Cell biology ,Fibroblast Growth Factors ,Liver ,PNAS Plus ,Exoribonucleases ,Hepatocytes ,030217 neurology & neurosurgery - Abstract
Whole-body metabolic homeostasis is tightly controlled by hormone-like factors with systemic or paracrine effects that are derived from nonendocrine organs, including adipose tissue (adipokines) and liver (hepatokines). Fibroblast growth factor 21 (FGF21) is a hormone-like protein, which is emerging as a major regulator of whole-body metabolism and has therapeutic potential for treating metabolic syndrome. However, the mechanisms that control FGF21 levels are not fully understood. Herein, we demonstrate that FGF21 production in the liver is regulated via a posttranscriptional network consisting of the CCR4–NOT deadenylase complex and RNA-binding protein tristetraprolin (TTP). In response to nutrient uptake, CCR4–NOT cooperates with TTP to degrade AU-rich mRNAs that encode pivotal metabolic regulators, including FGF21. Disruption of CCR4–NOT activity in the liver, by deletion of the catalytic subunit CNOT6L, increases serum FGF21 levels, which ameliorates diet-induced metabolic disorders and enhances energy expenditure without disrupting bone homeostasis. Taken together, our study describes a hepatic CCR4–NOT/FGF21 axis as a hitherto unrecognized systemic regulator of metabolism and suggests that hepatic CCR4–NOT may serve as a target for devising therapeutic strategies in metabolic syndrome and related morbidities.
- Published
- 2019
8. β(2)-Adrenergic receptor agonist induced hepatic steatosis in mice: modeling nonalcoholic fatty liver disease in hyperadrenergic states
- Author
-
Jeffrey L. Barnes, Falguni Das, Yun Shi, Jason Pizzini, Hanzhou Wang, Goutam Ghosh Choudhury, Amrita Kamat, Michael S. Katz, Parveez Ahamed Abdul Azees, Chih Ko Yeh, Mengwei Zang, and Susan T. Weintraub
- Subjects
0301 basic medicine ,Agonist ,medicine.medical_specialty ,Cirrhosis ,Physiology ,medicine.drug_class ,business.industry ,Endocrinology, Diabetes and Metabolism ,Fatty liver ,030209 endocrinology & metabolism ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Physiology (medical) ,Internal medicine ,Nonalcoholic fatty liver disease ,Lipogenesis ,medicine ,Steatosis ,Receptor ,business ,Cellular localization ,Research Article - Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of disorders ranging from hepatic steatosis [excessive accumulation of triglycerides (TG)] to nonalcoholic steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The molecular pathogenesis of steatosis and progression to more severe NAFLD remains unclear. Obesity and aging, two principal risk factors for NAFLD, are associated with a hyperadrenergic state. β-Adrenergic responsiveness in liver increases in animal models of obesity and aging, and in both is linked to increased hepatic expression of β(2)-adrenergic receptors (β(2)-ARs). We previously showed that in aging rodents intracellular signaling from elevated hepatic levels of β(2)-ARs may contribute to liver steatosis. In this study we demonstrate that injection of formoterol, a highly selective β(2)-AR agonist, to mice acutely results in hepatic TG accumulation. Further, we have sought to define the intrahepatic mechanisms underlying β(2)-AR mediated steatosis by investigating changes in hepatic expression and cellular localization of enzymes, transcription factors, and coactivators involved in processes of lipid accrual and disposition—and also functional aspects thereof—in livers of formoterol-treated animals. Our results suggest that β(2)-AR activation by formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, increased but incomplete β-oxidation of fatty acids with accumulation of potentially toxic long-chain acylcarnitine intermediates, and reduced TG secretion—all previously invoked as contributors to fatty liver disease. Experiments are ongoing to determine whether sustained activation of hepatic β(2)-AR signaling by formoterol might be utilized to model fatty liver changes occurring in hyperadrenergic states of obesity and aging, and thereby identify novel molecular targets for the prevention or treatment of NAFLD. NEW & NOTEWORTHY Results of our study suggest that β(2)-adrenergic receptor (β(2)-AR) activation by agonist formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, incomplete β-oxidation of fatty acids with accumulation of long-chain acylcarnitine intermediates, and reduced TG secretion. These findings may, for the first time, implicate a role for β(2)-AR responsive dysregulation of hepatic lipid metabolism in the pathogenetic processes underlying NAFLD in hyperadrenergic states such as obesity and aging.
- Published
- 2021
9. Alcohol Binge Drinking Selectively Stimulates Protein S-Glutathionylation in Aorta and Liver of ApoE−/− Mice
- Author
-
Kerstin Seidel, Xueping Wan, Mo Zhang, Yuxiang Zhou, Mengwei Zang, and Jingyan Han
- Subjects
0301 basic medicine ,Apolipoprotein E ,medicine.medical_specialty ,lcsh:Diseases of the circulatory (Cardiovascular) system ,Glutathione reductase ,Binge drinking ,Cardiovascular Medicine ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Protein S ,alcohol binge drinking ,aortic endothelial dysfunction ,03 medical and health sciences ,0302 clinical medicine ,cardiovascular disease ,Internal medicine ,medicine ,Glutaredoxin-1 ,Endothelial dysfunction ,Ethanol metabolism ,Glutathion-S-transferase-Pi ,Original Research ,ApoE −/− ,fatty liver ,biology ,business.industry ,Fatty liver ,ApoE−/− ,medicine.disease ,030104 developmental biology ,Endocrinology ,lcsh:RC666-701 ,biology.protein ,protein S-glutathionylation ,business ,Cardiology and Cardiovascular Medicine ,Oxidative stress - Abstract
Background: Binge drinking has become the most common and deadly pattern of excessive alcohol use in the United States, especially among younger adults. It is closely related to the increased risk of cardiovascular disease. Oxidative stress as a result of ethanol metabolism is the primary pathogenic factor for alcohol-induced end organ injury, but the role of protein S-glutathionylation—a reversible oxidative modification of protein cysteine thiol groups that mediates cellular actions by oxidants—in binge drinking-associated cardiovascular disease has not been explored. The present study defines the effect of alcohol binge drinking on the formation of protein S-glutathionylation in a mouse model of atherosclerosis.Methods and Results: To mimic the weekend binge drinking pattern in humans, ApoE deficient (ApoE−/−) mice on the Lieber-DeCarli liquid diet received ethanol or isocaloric maltose (as a control) gavages (5 g/kg/day, 2 consecutive days/week) for 6 weeks. The primary alcohol-targeted organs (liver, brain), and cardiovascular system (heart, aorta, lung) of these two groups of the mice were determined by measuring the protein S-glutathionylation levels and its regulatory enzymes including [Glutaredoxin1(Grx1), glutathione reductase (GR), glutathione-S-transferase Pi (GST-π)], as well as by assessing aortic endothelial function and liver lipid levels. Our results showed that binge drinking selectively stimulated protein S-glutathionylation in aorta, liver, and brain, which coincided with altered glutathionylation regulatory enzyme expression that is downregulated Grx1 and upregulated GST-π in aorta, massive upregulation of GST-π in liver, and no changes in Grx1 and GST-π in brain. Functionally, binge drinking induced aortic endothelial cell function, as reflected by increased aortic permeability and reduced flow-mediated vasodilation.Conclusions: This study is the first to provide in vivo evidence for differential effects of binge drinking on formation of protein S-glutathionylation and its enzymatic regulation system in major alcohol-target organs and cardiovascular system. The selective induction of protein S-glutathionylation in aorta and liver is associated with aortic endothelial dysfunction and fatty liver, which may be a potential redox mechanism for the increased risk of vascular disease in human binge-drinkers.
- Published
- 2021
- Full Text
- View/download PDF
10. Aging aggravates alcoholic liver injury and fibrosis in mice by downregulating sirtuin 1 expression
- Author
-
Yong Mei Li, Zhou Zhou, Pal Pacher, Bin Gao, Mengwei Zang, Zoltán Varga, Mingjiang Xu, Hua Wang, Shi Yin, Teresa Ramirez, Partha Mukhopadhyay, and Dechun Feng
- Subjects
Male ,0301 basic medicine ,Aging ,medicine.medical_specialty ,Alcoholic liver disease ,Down-Regulation ,Article ,Binge Drinking ,Mice ,03 medical and health sciences ,Sirtuin 1 ,Downregulation and upregulation ,Fibrosis ,Internal medicine ,Hepatic Stellate Cells ,medicine ,Animals ,Liver Diseases, Alcoholic ,Mice, Knockout ,Liver injury ,Hepatology ,biology ,Fatty liver ,medicine.disease ,Liver Regeneration ,Mice, Inbred C57BL ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Immunology ,biology.protein ,Hepatic stellate cell ,Female ,Steatohepatitis - Abstract
Background & Aims Aging is known to exacerbate the progression of alcoholic liver disease (ALD), but the underlying mechanisms remain obscure. The aim of this study was to use a chronic plus binge ethanol feeding model in mice to evaluate the effects of aging on alcohol-induced liver injury. Methods C57BL/6 mice were subjected to short-term (10days) ethanol plus one binge or long-term (8weeks) ethanol plus multiple binges of ethanol. Liver injury and fibrosis were determined. Hepatic stellate cells (HSCs) were isolated and used in in vitro studies. Results Middle-aged (12–14months) and old-aged (>16months) mice were more susceptible to liver injury, inflammation, and oxidative stress induced by short-term plus one binge or long-term plus multiple binges of ethanol feeding when compared to young (8–12weeks) mice. Long-term plus multiple binges of ethanol feeding induced greater liver fibrosis in middle-aged mice than that in young mice. Hepatic expression of sirtuin 1 (SIRT1) protein was downregulated in the middle-aged mice compared to young mice. Restoration of SIRT1 expression via the administration of adenovirus-SIRT1 vector ameliorated short-term plus binge ethanol-induced liver injury and fibrosis in middle-aged mice. HSCs isolated from middle-aged mice expressed lower levels of SIRT1 protein and were more susceptible to spontaneous activation in in vitro culture than those from young mice. Overexpression of SIRT1 reduced activation of HSCs from middle-aged mice in vitro with downregulation of PDGFR-α and c-Myc, while deletion of SIRT1 activated HSCs isolated from young mice in vitro . Finally, HSC-specific SIRT1 knockout mice were more susceptible to long-term chronic-plus-multiple binges of ethanol-induced liver fibrosis with upregulation of PDGFR-α expression. Conclusions Aging exacerbates ALD in mice through the downregulation of SIRT1 in hepatocytes and HSCs. Activation of SIRT1 may serve as a novel target for the treatment of ALD. Lay summary Aged mice are more susceptible to alcohol-induced liver injury and fibrosis, which is, at least in part, due to lower levels of sirtuin 1 protein in hepatocytes and hepatic stellate cells. Our findings suggest that sirtuin 1 activators may have beneficial effects for the treatment of alcoholic liver disease in aged patients.
- Published
- 2017
11. The redox mechanism for vascular barrier dysfunction associated with metabolic disorders: Glutathionylation of Rac1 in endothelial cells
- Author
-
Naomi M. Hamburg, Richard A. Cohen, Mengwei Zang, Markus Bachschmid, Di Shao, Xiaoyan Yin, Yvonne M. W. Janssen-Heininger, Reiko Matsui, Yosuke Watanabe, Jingyan Han, Francesca Seta, and Robert M. Weisbrod
- Subjects
Male ,rac1 GTP-Binding Protein ,0301 basic medicine ,Apolipoprotein E ,Clinical Biochemistry ,Gene Expression ,Vascular permeability ,medicine.disease_cause ,Biochemistry ,Mice ,lcsh:QH301-705.5 ,Aorta ,Mice, Knockout ,lcsh:R5-920 ,Actin cytoskeleton ,Endothelial barrier function ,Glutathione ,Cell biology ,Endothelial stem cell ,lcsh:Medicine (General) ,Oxidation-Reduction ,Research Paper ,RAC1 ,Oxidative phosphorylation ,Biology ,Cell Line ,Capillary Permeability ,Adherens junction ,ApoE-deficient mice ,03 medical and health sciences ,Apolipoproteins E ,Metabolic Diseases ,Small Rho GTPase Rac1 ,Stress, Physiological ,medicine ,Animals ,Humans ,Glutaredoxin-1 ,Cysteine ,Glutaredoxins ,Organic Chemistry ,Endothelial Cells ,030104 developmental biology ,lcsh:Biology (General) ,Mutation ,Endothelium, Vascular ,Protein S-glutathionylation ,Protein Processing, Post-Translational ,Oxidative stress - Abstract
Background Oxidative stress is implicated in increased vascular permeability associated with metabolic disorders, but the underlying redox mechanism is poorly defined. S-glutathionylation, a stable adduct of glutathione with protein sulfhydryl, is a reversible oxidative modification of protein and is emerging as an important redox signaling paradigm in cardiovascular physiopathology. The present study determines the role of protein S-glutathionylation in metabolic stress-induced endothelial cell permeability. Methods and results In endothelial cells isolated from patients with type-2 diabetes mellitus, protein S-glutathionylation level was increased. This change was also observed in aortic endothelium in ApoE deficient (ApoE-/-) mice fed on Western diet. Metabolic stress-induced protein S-glutathionylation in human aortic endothelial cells (HAEC) was positively correlated with elevated endothelial cell permeability, as reflected by disassembly of cell-cell adherens junctions and cortical actin structures. These impairments were reversed by adenoviral overexpression of a specific de-glutathionylation enzyme, glutaredoxin-1 in cultured HAECs. Consistently, transgenic overexpression of human Glrx-1 in ApoE-/- mice fed the Western diet attenuated endothelial protein S-glutathionylation, actin cytoskeletal disorganization, and vascular permeability in the aorta. Mechanistically, glutathionylation and inactivation of Rac1, a small RhoGPase, were associated with endothelial hyperpermeability caused by metabolic stress. Glutathionylation of Rac1 on cysteine 81 and 157 located adjacent to guanine nucleotide binding site was required for the metabolic stress to inhibit Rac1 activity and promote endothelial hyperpermeability. Conclusions Glutathionylation and inactivation of Rac1 in endothelial cells represent a novel redox mechanism of vascular barrier dysfunction associated with metabolic disorders., Graphical abstract fx1, Highlights • In metabolically stressed endothelial cells, protein S-glutathionylation is elevated. • glutaredoxin-1 diminishes protein S-glutathionylation and preserves aortic barrier function. • Pharmacological inhibition of Rac1 abrogates Glrx1-mediated barrier protection. • Glutathionylation of Rac1 is associated with a defect in Rac1 activation status.
- Published
- 2016
12. AMPK Activation by Metformin Suppresses Abnormal Extracellular Matrix Remodeling in Adipose Tissue and Ameliorates Insulin Resistance in Obesity
- Author
-
Ting Luo, Qin Yang, Alex Sherban, Mengwei Zang, Jingyan Han, Yun Yan, Allison Nocon, Jessica L. Fry, Xianliang Rui, Bingbing Jiang, Qifu Li, and X. Julia Xu
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,FGF21 ,Adipose Tissue, White ,Endocrinology, Diabetes and Metabolism ,Adipose tissue ,White adipose tissue ,AMP-Activated Protein Kinases ,In Vitro Techniques ,Transforming Growth Factor beta1 ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,AMP-activated protein kinase ,Fibrosis ,Internal medicine ,Adipocyte ,Internal Medicine ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Obesity ,Myofibroblasts ,Cells, Cultured ,biology ,Chemistry ,AMPK ,Stromal vascular fraction ,medicine.disease ,Metformin ,Extracellular Matrix ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Adipose Tissue ,biology.protein ,Collagen ,Insulin Resistance ,Obesity Studies ,Signal Transduction - Abstract
Fibrosis is emerging as a hallmark of metabolically dysregulated white adipose tissue (WAT) in obesity. Although adipose tissue fibrosis impairs adipocyte plasticity, little is known about how aberrant extracellular matrix (ECM) remodeling of WAT is initiated during the development of obesity. Here we show that treatment with the antidiabetic drug metformin inhibits excessive ECM deposition in WAT of ob/ob mice and mice with diet-induced obesity, as evidenced by decreased collagen deposition surrounding adipocytes and expression of fibrotic genes including the collagen cross-linking regulator LOX. Inhibition of interstitial fibrosis by metformin is likely attributable to the activation of AMPK and the suppression of transforming growth factor-β1 (TGF-β1)/Smad3 signaling, leading to enhanced systemic insulin sensitivity. The ability of metformin to repress TGF-β1-induced fibrogenesis is abolished by the dominant negative AMPK in primary cells from the stromal vascular fraction. TGF-β1-induced insulin resistance is suppressed by AMPK agonists and the constitutively active AMPK in 3T3L1 adipocytes. In omental fat depots of obese humans, interstitial fibrosis is also associated with AMPK inactivation, TGF-β1/Smad3 induction, aberrant ECM production, myofibroblast activation, and adipocyte apoptosis. Collectively, integrated AMPK activation and TGF-β1/Smad3 inhibition may provide a potential therapeutic approach to maintain ECM flexibility and combat chronically uncontrolled adipose tissue expansion in obesity.
- Published
- 2016
13. Organ-specific alterations in circadian genes by vertical sleeve gastrectomy in an obese diabetic mouse model
- Author
-
Xiaolong Zhao, Zhiyu Hu, Jingjing Zhu, Qiyuan Yao, Meng Wang, Rong Hua, Wanzhu Jin, Mengwei Zang, Qinghua Wang, Bo Xu, Wenjuan Liu, Yikai Shao, Yiming Li, Zhaoyun Zhang, Yeping Yang, Jae Bum Kim, and Qiwei Shen
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Sleeve gastrectomy ,Multidisciplinary ,business.industry ,medicine.medical_treatment ,Diabetic mouse ,Article ,03 medical and health sciences ,030104 developmental biology ,Endocrinology ,Text mining ,Internal medicine ,Organ specific ,medicine ,Circadian rhythm ,business ,Gene - Published
- 2017
14. DEP domain-containing mTOR-interacting protein suppresses lipogenesis and ameliorates hepatic steatosis and acute-on-chronic liver injury in alcoholic liver disease
- Author
-
Pal Pacher, Yu Li, Mengwei Zang, Jian-Gao Fan, Feng Liu, Jinyan Han, Bingbing Jiang, Bin Gao, Allison Nocon, Donghwan Lee, Leonard Guarente, Alex Sherban, Nicolas Musi, Feng Shen, Hanqing Chen, Hua Wang, Mingjiang Xu, Xianliang Rui, Amrita Kamat, Na Li, Farnaz Keyhani-Nejad, and Massachusetts Institute of Technology. Department of Biology
- Subjects
0301 basic medicine ,Liver injury ,medicine.medical_specialty ,Alcoholic liver disease ,Hepatology ,Fatty liver ,P70-S6 Kinase 1 ,mTORC1 ,medicine.disease ,DEPTOR ,03 medical and health sciences ,030104 developmental biology ,Endocrinology ,Internal medicine ,Lipogenesis ,medicine ,Alcoholic fatty liver ,biological phenomena, cell phenomena, and immunity - Abstract
Alcoholic liver disease (ALD) is characterized by lipid accumulation and liver injury. However, how chronic alcohol consumption causes hepatic lipid accumulation remains elusive. The present study demonstrates that activation of the mechanistic target of rapamycin complex 1 (mTORC1) plays a causal role in alcoholic steatosis, inflammation, and liver injury. Chronic-plus-binge ethanol feeding led to hyperactivation of mTORC1, as evidenced by increased phosphorylation of mTOR and its downstream kinase S6 kinase 1 (S6K1) in hepatocytes. Aberrant activation of mTORC1 was likely attributed to the defects of the DEP domain-containing mTOR-interacting protein (DEPTOR) and the nicotinamide adenine dinucleotide-dependent deacetylase sirtuin 1 (SIRT1) in the liver of chronic-plus-binge ethanol-fed mice and in the liver of patients with ALD. Conversely, adenoviral overexpression of hepatic DEPTOR suppressed mTORC1 signaling and ameliorated alcoholic hepatosteatosis, inflammation, and acute-on-chronic liver injury. Mechanistically, the lipid-lowering effect of hepatic DEPTOR was attributable to decreased proteolytic processing, nuclear translocation, and transcriptional activity of the lipogenic transcription factor sterol regulatory element-binding protein-1 (SREBP-1). DEPTOR-dependent inhibition of mTORC1 also attenuated alcohol-induced cytoplasmic accumulation of the lipogenic regulator lipin 1 and prevented alcohol-mediated inhibition of fatty acid oxidation. Pharmacological intervention with rapamycin alleviated the ability of alcohol to up-regulate lipogenesis, to down-regulate fatty acid oxidation, and to induce steatogenic phenotypes. Chronic-plus-binge ethanol feeding led to activation of SREBP-1 and lipin 1 through S6K1-dependent and independent mechanisms. Furthermore, hepatocyte-specific deletion of SIRT1 disrupted DEPTOR function, enhanced mTORC1 activity, and exacerbated alcoholic fatty liver, inflammation, and liver injury in mice. Conclusion The dysregulation of SIRT1-DEPTOR-mTORC1 signaling is a critical determinant of ALD pathology; targeting SIRT1 and DEPTOR and selectively inhibiting mTORC1-S6K1 signaling may have therapeutic potential for treating ALD in humans. (Hepatology 2018).
- Published
- 2018
15. New Insight Into Metformin Action: Regulation of ChREBP and FOXO1 Activities in Endothelial Cells
- Author
-
Dara J. Watkins, Xiaoyu Li, Karen Kover, Yun Yan, Kathyrin Jackson, Mengwei Zang, Daniel P. Heruth, Mark A. Clements, and Wayne V. Moore
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Cell Cycle Proteins ,Nerve Tissue Proteins ,FOXO1 ,Biology ,Rats, Sprague-Dawley ,Mice ,Cytosol ,Thioredoxins ,Endocrinology ,Insulin resistance ,Internal medicine ,medicine ,Animals ,Humans ,Endothelial dysfunction ,Nuclear protein ,Carbohydrate-responsive element-binding protein ,Molecular Biology ,Aorta ,Original Research ,Cell Nucleus ,Inflammation ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Forkhead Box Protein O1 ,Adenylate Kinase ,Endothelial Cells ,Nuclear Proteins ,Forkhead Transcription Factors ,General Medicine ,Protein kinase inhibitor ,medicine.disease ,Metformin ,Rats ,Oxidative Stress ,Diabetes Mellitus, Type 1 ,Carrier Proteins ,Reactive Oxygen Species ,Oxidation-Reduction ,TXNIP ,Transcription Factors ,medicine.drug - Abstract
Metformin has been considered a potential adjunctive therapy in treating poorly controlled type 1 diabetes with obesity and insulin resistance, owing to its potent effects on improving insulin sensitivity. However, the underlying mechanism of metformin's vascular protective effects remains obscure. Thioredoxin-interacting protein (TXNIP), a key regulator of cellular redox state induced by high-glucose concentration, decreases thioredoxin reductase activity and mediates apoptosis induced by oxidative stress. Here we report that high glucose-induced endothelial dysfunction is associated with induction of TXNIP expression in primary human aortic endothelial cells exposed to high-glucose conditions, whereas the metformin treatment suppresses high-glucose-induced TXNIP expression at mRNA and protein levels. We further show that metformin decreases the high-glucose-stimulated nuclear entry rate of two transcription factors, carbohydrate response element-binding protein (ChREBP) and forkhead box O1 (FOXO1), as well as their recruitment on the TXNIP promoter. An AMP-activated protein kinase inhibitor partially compromised these metformin effects. Our data suggest that endothelial dysfunction resulting from high-glucose concentrations is associated with TXNIP expression. Metformin down-regulates high-glucose-induced TXNIP transcription by inactivating ChREBP and FOXO1 in endothelial cells, partially through AMP-activated protein kinase activation.
- Published
- 2015
16. Thioredoxin-interacting protein promotes high-glucose-induced macrovascular endothelial dysfunction
- Author
-
Dara J. Watkins, Luke G. He, Yun Yan, Xiaoyu Li, Yanchun Guo, Karen Kover, Mengwei Zang, Wayne V. Moore, Mark A. Clements, and Daniel P. Heruth
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Thioredoxin-Interacting Protein ,Biophysics ,Aortic Diseases ,Cell Cycle Proteins ,030204 cardiovascular system & hematology ,Biology ,Biochemistry ,Article ,Nitric oxide ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,medicine ,Glucose homeostasis ,Animals ,Endothelial dysfunction ,Carbohydrate-responsive element-binding protein ,Molecular Biology ,Cell Biology ,medicine.disease ,Rats ,Vascular endothelial growth factor ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 1 ,Glucose ,chemistry ,Cancer research ,Endothelium, Vascular ,Carrier Proteins ,Reactive Oxygen Species ,TXNIP ,Diabetic Angiopathies - Abstract
Thioredoxin-interacting protein (TXNIP) emerges as a central regulator for glucose homeostasis, which goes awry in diabetic subjects. Endothelial dysfunction is considered the earliest detectable stage of cardiovascular disease (CVD), a major complication of diabetes. Here, we hypothesize that TXNIP may promote endothelial dysfunction seen in Type 1 diabetes mellitus (T1D). Using a T1D-like rat model, we found that diabetic rats showed significantly higher TXNIP mRNA and protein levels in peripheral blood, compared to their non-diabetic counterparts. Those changes were accompanied by decreased production of nitric oxide (NO) and vascular endothelial growth factor (VEGF), concurrent with increased expression of reactive oxygen species (ROS) and vascular cell adhesion molecule 1 (VCAM-1) in the aortic endothelium. In addition, TXNIP overexpression in primary human aortic endothelial cells (HAECs) induced by either high glucose or overexpression of carbohydrate response element binding protein (ChREBP), a major transcriptional activator of TXNIP, promoted early apoptosis and impaired NO bioactivity. The correlation between TXNIP expression levels and endothelial dysfunction suggests that TXNIP may be a potential biomarker for vascular complications in T1D patients.
- Published
- 2017
17. SIRT1 Deacetylates and Inhibits SREBP-1C Activity in Regulation of Hepatic Lipid Metabolism*
- Author
-
Zhen Xiao, Zachary Smith, Cheng Ming Chiang, Timothy D. Veenstra, Ji Miao, Dong-Hyun Kim, Mengwei Zang, Bhaskar Ponugoti, Jongsook Kim Kemper, and Shwu Yuan Wu
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,medicine.medical_treatment ,digestive system ,Biochemistry ,Mass Spectrometry ,Mice ,Transactivation ,Sirtuin 1 ,Internal medicine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Insulin ,Molecular Biology ,Regulation of gene expression ,Mice, Inbred BALB C ,biology ,Activator (genetics) ,Fatty liver ,food and beverages ,Cell Biology ,medicine.disease ,Lipids ,Metabolism ,Endocrinology ,Gene Expression Regulation ,Liver ,Acetylation ,COS Cells ,Lipogenesis ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Sterol Regulatory Element Binding Protein 1 - Abstract
The SIRT1 deacetylase inhibits fat synthesis and stimulates fat oxidation in response to fasting, but the underlying mechanisms remain unclear. Here we report that SREBP-1c, a key lipogenic activator, is an in vivo target of SIRT1. SIRT1 interaction with SREBP-1c was increased during fasting and decreased upon feeding, and consistently, SREBP-1c acetylation levels were decreased during fasting in mouse liver. Acetylated SREBP-1c levels were also increased in HepG2 cells treated with insulin and glucose to mimic feeding conditions, and down-regulation of p300 by siRNA decreased the acetylation. Depletion of hepatic SIRT1 by adenoviral siRNA increased acetylation of SREBP-1c with increased lipogenic gene expression. Tandem mass spectrometry and mutagenesis studies revealed that SREBP-1c is acetylated by p300 at Lys-289 and Lys-309. Mechanistic studies using acetylation-defective mutants showed that SIRT1 deacetylates and inhibits SREBP-1c transactivation by decreasing its stability and its occupancy at the lipogenic genes. Remarkably, SREBP-1c acetylation levels were elevated in diet-induced obese mice, and hepatic overexpression of SIRT1 or treatment with resveratrol, a SIRT1 activator, daily for 1 week decreased acetylated SREBP-1c levels with beneficial functional outcomes. These results demonstrate an intriguing connection between elevated SREBP-1c acetylation and increased lipogenic gene expression, suggesting that abnormally elevated SREBP-1c acetylation increases SREBP-1c lipogenic activity in obese mice. Reducing acetylation of SREBP-1c by targeting SIRT1 may be useful for treating metabolic disorders, including fatty liver, obesity, and type II diabetes.
- Published
- 2010
18. AMPK as a metabolic tumor suppressor: control of metabolism and cell growth
- Author
-
Mengwei Zang, Wen Guo, and Zhijun Luo
- Subjects
Cancer Research ,medicine.medical_specialty ,mTORC1 ,medicine.disease_cause ,Article ,AMP-Activated Protein Kinase Kinases ,Neoplasms ,Internal medicine ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Beta oxidation ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Metabolic Syndrome ,biology ,business.industry ,AMPK ,General Medicine ,Fatty acid synthase ,Endocrinology ,Oncology ,Cancer cell ,biology.protein ,Cancer research ,Signal transduction ,Energy Metabolism ,Carcinogenesis ,business ,Protein Kinases ,Signal Transduction - Abstract
AMPK is an evolutionarily conserved fuel-sensing enzyme that is activated in shortage of energy and suppressed in its surfeit. AMPK activation stimulates fatty acid oxidation, enhances insulin sensitivity, alleviates hyperglycemia and hyperlipidemia, and inhibits proinflammatory changes. Thus, AMPK is a well-received therapeutic target for metabolic syndrome and Type 2 diabetes. Recent studies indicate that AMPK plays a role in linking metabolic syndrome and cancer. AMPK is an essential mediator of the tumor suppressor LKB1 and could be suppressed in cancer cells containing loss-of-function mutations of LKB1 or containing active mutations of B-Raf, or in cancers associated with metabolic syndrome. The activation of AMPK reprograms cellular metabolism and enforces metabolic checkpoints by acting on mTORC1, p53, fatty acid synthase and other molecules for regulating cell growth and metabolism. In keeping with in vitro studies, recent epidemiological studies indicate that the incidence of cancer is reduced in Type 2 diabetes treated with metformin, an AMPK activator. Thus, AMPK is emerging as an interesting metabolic tumor suppressor and a promising target for cancer prevention and therapy.
- Published
- 2010
19. Overnutrition and maternal obesity in sheep pregnancy alter the JNK‐IRS‐1 signaling cascades and cardiac function in the fetal heart
- Author
-
Mark J. Nijland, Jun Ren, Gavin B. Lawlis, Peter W. Nathanielsz, Heng Ma, Mengwei Zang, Yuanda Li, Jingying Wang, Stephen P. Ford, Chao Tong, Hanying Zhang, and Ji Li
- Subjects
Cardiac function curve ,medicine.medical_specialty ,MAP Kinase Signaling System ,medicine.medical_treatment ,Immunoblotting ,Biochemistry ,Research Communications ,Fetal Heart ,Overnutrition ,Insulin resistance ,Pregnancy ,Internal medicine ,Genetics ,medicine ,Animals ,Immunoprecipitation ,Insulin ,Obesity ,Phosphorylation ,Molecular Biology ,Fetus ,Sheep ,biology ,JNK Mitogen-Activated Protein Kinases ,AMPK ,medicine.disease ,Insulin receptor ,Phenotype ,Endocrinology ,Insulin Receptor Substrate Proteins ,biology.protein ,Female ,Insulin Resistance ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,Biotechnology - Abstract
Maternal obesity in pregnancy predisposes offspring to insulin resistance and associated cardiovascular disease. Here, we used a well-established sheep model to investigate the effects of maternal obesity on cardiac functions. Multiparous ewes were assigned to a control (CON) diet [100% of National Research Council (NRC) recommendations] or an obesogenic (OB) diet (150% of NRC recommendations) from 60 d before conception to necropsy on d 135 of pregnancy. Fetal blood glucose and insulin were increased (P
- Published
- 2010
20. Fibroblast growth factor 21 improves hepatic insulin sensitivity by inhibiting mammalian target of rapamycin complex 1 in mice
- Author
-
Qi Gong, Qingning Liang, Yu Li, Yong Liu, Aimin Xu, Jing Gao, Mengwei Zang, Yamei Han, Yu Wang, Xuqing Chen, Hui Xiao, Dewei Ye, Feifei Zhang, Y. Eugene Chin, Zhimin Hu, Lei Shi, Feifan Guo, Xin Chen, Aoyuan Cui, and Haoyang Jiang
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Sucrose ,FGF21 ,medicine.medical_treatment ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Diet, High-Fat ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,medicine ,Glucose homeostasis ,Animals ,Insulin ,Glycogen synthase ,Protein kinase B ,Klotho Proteins ,Mice, Knockout ,Hepatology ,biology ,TOR Serine-Threonine Kinases ,Membrane Proteins ,medicine.disease ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,Insulin receptor ,030104 developmental biology ,Endocrinology ,Liver ,030220 oncology & carcinogenesis ,Multiprotein Complexes ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Insulin Resistance ,Glycogen - Abstract
Among the 22 fibroblast growth factors (FGFs), FGF21 has now emerged as a key metabolic regulator. However, the mechanism whereby FGF21 mediates its metabolic actions per se remains largely unknown. Here, we show that FGF21 represses mammalian target of rapamycin complex 1 (mTORC1) and improves insulin sensitivity and glycogen storage in a hepatocyte-autonomous manner. Administration of FGF21 in mice inhibits mTORC1 in the liver, whereas FGF21-deficient mice display pronounced insulin-stimulated mTORC1 activation and exacerbated hepatic insulin resistance (IR). FGF21 inhibits insulin- or nutrient-stimulated activation of mTORC1 to enhance phosphorylation of Akt in HepG2 cells at both normal and IR condition. TSC1 deficiency abrogates FGF21-mediated inhibition of mTORC1 and augmentation of insulin signaling and glycogen synthesis. Strikingly, hepatic βKlotho knockdown or hepatic hyperactivation of mTORC1/ribosomal protein S6 kinase 1 abrogates hepatic insulin-sensitizing and glycemic-control effects of FGF21 in diet-induced insulin-resistant mice. Moreover, FGF21 improves methionine- and choline-deficient diet-induced steatohepatitis. Conclusions: FGF21 acts as an inhibitor of mTORC1 to control hepatic insulin action and maintain glucose homeostasis, and mTORC1 inhibition by FGF21 has the therapeutic potential for treating IR and type 2 diabetes. (Hepatology 2016;64:425-438)
- Published
- 2015
21. mTORC1 Inhibition Reduces ER Stress and Ameliorates Alcoholic Liver Injury
- Author
-
Bin Gao, Mengwei Zang, Farhan Ladhani, and Shan-Shan Li
- Subjects
Liver injury ,medicine.medical_specialty ,business.industry ,mTORC1 ,medicine.disease ,Biochemistry ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Unfolded protein response ,business ,Molecular Biology ,Biotechnology - Published
- 2015
22. The Thromboxane A 2 Receptor Antagonist S18886 Prevents Enhanced Atherogenesis Caused by Diabetes Mellitus
- Author
-
Antonio J. Cayatte, Gilbert Lavielle, Adriana Zuccollo, Bingbing Jiang, Karlene A. Maitland-Toolan, Richard A. Cohen, Roberto Mastroianni, Shanqin Xu, Chaomei Shi, Mengwei Zang, Jennifer Oliver-Krasinski, Stefano Corda, Robert M. Weisbrod, and Tony Verbeuren
- Subjects
Male ,medicine.medical_specialty ,Apolipoprotein B ,Thromboxane ,Naphthalenes ,Receptors, Thromboxane A2, Prostaglandin H2 ,Diabetes Mellitus, Experimental ,Lesion ,Mice ,Thromboxane A2 ,chemistry.chemical_compound ,Apolipoproteins E ,Physiology (medical) ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Crosses, Genetic ,Mice, Knockout ,biology ,Cholesterol ,business.industry ,Antagonist ,Atherosclerosis ,medicine.disease ,Streptozotocin ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,biology.protein ,Female ,Propionates ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Diabetic Angiopathies ,medicine.drug - Abstract
Background— S18886 is an orally active thromboxane A 2 (TXA 2 ) receptor (TP) antagonist in clinical development for use in secondary prevention of thrombotic events in cardiovascular disease. We previously showed that S18886 inhibits atherosclerosis in apolipoprotein E–deficient (apoE −/− ) mice by a mechanism independent of platelet-derived TXA 2 . Atherosclerosis is accelerated by diabetes and is associated with increased TXA 2 and other eicosanoids that stimulate TP. The purpose of this study was to determine whether S18886 lessens the enhanced atherogenesis in diabetic apoE −/− mice. Methods and Results— Diabetes mellitus was induced in apoE −/− mice with streptozotocin and was treated or not with S18886 (5 mg · kg −1 · d −1 ). After 6 weeks, aortic lesion area was increased >4-fold by diabetes in apoE −/− mice, associated with similar increases in serum glucose and cholesterol. S18886 largely prevented the diabetes-related increase in lesion area without affecting the hyperglycemia or hypercholesterolemia. S18886 prevented deterioration of endothelial function and endothelial nitric oxide synthase expression, as well as increases in intimal markers of inflammation associated with diabetes. In human aortic endothelial cells in culture, S18886 also prevented the induction of vascular cell adhesion molecule-1 and prevented the decrease in endothelial nitric oxide synthase expression caused by high glucose. Conclusions— The TP antagonist inhibits inflammation and accelerated atherogenesis caused by diabetes, most likely by counteracting effects on endothelial function and adhesion molecule expression of eicosanoids stimulated by the diabetic milieu.
- Published
- 2005
23. AMP-activated Protein Kinase Is Required for the Lipid-lowering Effect of Metformin in Insulin-resistant Human HepG2 Cells
- Author
-
Peter Brecher, Mengwei Zang, Adriana Zuccollo, Daisuke Nagata, Haya Herscovitz, Neil B. Ruderman, Xiuyun Hou, Kenneth Walsh, and Richard A. Cohen
- Subjects
medicine.medical_specialty ,Recombinant Fusion Proteins ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Biochemistry ,Glycogen Synthase Kinase 3 ,Insulin resistance ,AMP-activated protein kinase ,Multienzyme Complexes ,GSK-3 ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Internal medicine ,medicine ,Humans ,Hypoglycemic Agents ,Insulin ,Protein kinase A ,Molecular Biology ,Protein kinase B ,Glycogen Synthase Kinase 3 beta ,biology ,Chemistry ,AMPK ,Lipid metabolism ,Cell Biology ,Lipid Metabolism ,medicine.disease ,Metformin ,Enzyme Activation ,Protein Subunits ,Glucose ,Endocrinology ,Liver ,biology.protein ,Insulin Resistance ,Proto-Oncogene Proteins c-akt ,Acetyl-CoA Carboxylase ,medicine.drug - Abstract
The antidiabetic drug metformin stimulates AMP-activated protein kinase (AMPK) activity in the liver and in skeletal muscle. To better understand the role of AMPK in the regulation of hepatic lipids, we studied the effect of metformin on AMPK and its downstream effector, acetyl-CoA carboxylase (ACC), as well as on lipid content in cultured human hepatoma HepG2 cells. Metformin increased Thr-172 phosphorylation of the alpha subunit of AMPK in a dose- and time-dependent manner. In parallel, phosphorylation of ACC at Ser-79 was increased, which was consistent with decreasing ACC activity. Intracellular triacylglycerol and cholesterol contents were also decreased. These effects of metformin were mimicked or completely abrogated by adenoviral-mediated expression of a constitutively active AMPKalpha or a kinase-inactive AMPKalpha, respectively. An insulin-resistant state was induced by exposing cells to 30 mm glucose as indicated by decreased phosphorylation of Akt and its downstream effector, glycogen synthase kinase 3alpha/beta. Under these conditions, the phosphorylation of AMPK and ACC was also decreased, and the level of hepatocellular triacylglycerols increased. The inhibition of AMPK and the accumulation of lipids caused by high glucose concentrations were prevented either by metformin or by expressing the constitutively active AMPKalpha. The kinase-inactive AMPKalpha increased lipid content and blocked the ability of metformin to decrease lipid accumulation caused by high glucose concentrations. Taken together, these results indicate that AMPKalpha negatively regulates ACC activity and hepatic lipid content. Inhibition of AMPK may contribute to lipid accumulation induced by high concentrations of glucose associated with insulin resistance. Metformin lowers hepatic lipid content by activating AMPK, thereby mediating beneficial effects in hyperglycemia and insulin resistance.
- Published
- 2004
24. Spatial Approximation between Two Residues in the Mid-region of Secretin and the Amino Terminus of Its Receptor
- Author
-
Maoqing Dong, Delia I. Pinon, Laurence J. Miller, Terry P. Lybrand, Mengwei Zang, and Zhijun Li
- Subjects
chemistry.chemical_classification ,Affinity labeling ,Photoaffinity labeling ,Chemistry ,Stereochemistry ,Peptide ,Cell Biology ,Biochemistry ,Transmembrane domain ,Secretin receptor ,GABBR2 ,GABBR1 ,Receptor ,Molecular Biology - Abstract
Photoaffinity labeling of receptors by bound agonists can provide important spatial constraints for molecular modeling of activated receptor complexes. Secretin is a 27-residue peptide hormone with a diffuse pharmacophoric domain that binds to the secretin receptor, a prototypic member of the Class B family of G protein-coupled receptors. In this work, we have developed, characterized, and applied two new photolabile probes for this receptor, with sites for covalent attachment in peptide positions 12 and 14, surrounding the previously most informative site of affinity labeling of this receptor. The [Tyr10,(BzBz)Lys12]rat secretin-27 probe covalently labeled receptor residue Val6, whereas the [Tyr10,(BzBz)Lys14]rat secretin-27 probe labeled receptor residue Pro38. When combined with previous photoaffinity labeling data, there are now seven independent sets of constraints distributed throughout the peptide and receptor amino-terminal domain that can be used together to generate a new molecular model of the ligand-occupied secretin receptor. The aminoterminal domain of this receptor presented a stable platform for peptide ligand interaction, with the amino terminus of the peptide hormone extended toward the transmembrane helix domain of the receptor. This provides clear insights into the molecular basis of natural ligand binding and supplies testable hypotheses regarding the molecular basis of activation of this receptor.
- Published
- 2003
25. Phosphorylation of 338SSYY341 Regulates Specific Interaction between Raf-1 and MEK1
- Author
-
Zhijun Luo, Rong Wen, Christine A. Waelde, Mengwei Zang, and Xiaoqin Xiang
- Subjects
Recombinant Fusion Proteins ,MAP Kinase Kinase 1 ,Context (language use) ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,Biochemistry ,Cell Line ,Epitopes ,Serine ,medicine ,Humans ,Amino Acid Sequence ,c-Raf ,Phosphorylation ,Kinase activity ,Molecular Biology ,Mitogen-Activated Protein Kinase Kinases ,chemistry.chemical_classification ,Mutation ,Binding Sites ,Kinase ,Cell Biology ,Amino acid ,Enzyme Activation ,Proto-Oncogene Proteins c-raf ,Protein kinase domain ,chemistry ,Mutagenesis, Site-Directed ,Tetradecanoylphorbol Acetate ,Tyrosine ,Protein Binding - Abstract
The present study characterizes the interaction between the Raf-1 kinase domain and MEK1 and examines whether the magnitude of their interaction correlates to the ability of Raf to phosphorylate MEK1. Here we show that the minimal domain required for the Raf kinase activity starts from tryptophan 342. Maximal binding of the Raf kinase domain to MEK1 and its kinase activity are achieved upon phosphorylation of the region (338)SSYY(341) in response to 4beta-12-O-tetradecanoylphorbol-13-acetate (TPA), or mutation of Y340Y341 to aspartic acids. Conversely, the TPA-stimulated MEK binding and kinase activity are diminished when this region is deleted or Ser(338) and Ser(339) are mutated to alanines. We also show that the integrity of the Raf ATP-binding site is necessary for the interaction between Raf-1 and MEK1. Furthermore, two MEK-binding sites are identified; the first is localized between amino acids 325 and 349, and the second is within the region between amino acids 350 and 648. Separately, the binding of each site to MEK1 is weak, but in a cis context, they give rise to a much stronger association, which can be further stimulated by TPA. Finally, we find that tryptophan 342, which is conserved among the Raf family and other protein kinases, is essential for the Ser(338) phosphorylation of the full-length Raf and its binding to MEK1. Taken together, our results indicate that the phosphorylation of Ser(338) and Tyr(341) on Raf exerts an important effect on reconfiguring the two MEK-binding sites. As a result, these two sites coordinate to form a high affinity MEK-binding epitope, leading to a marked increase in Raf kinase activity.
- Published
- 2002
26. Interaction among Four Residues Distributed through the Secretin Pharmacophore and a Focused Region of the Secretin Receptor Amino Terminus
- Author
-
Maoqing Dong, Delia I. Pinon, Terry P. Lybrand, Mengwei Zang, Zhijun Li, and Laurence J. Miller
- Subjects
Models, Molecular ,Protein Conformation ,Molecular Sequence Data ,Peptide ,Biology ,Protein Structure, Secondary ,Receptors, G-Protein-Coupled ,Receptors, Gastrointestinal Hormone ,Secretin ,Endocrinology ,Protein structure ,Animals ,Amino Acid Sequence ,Binding site ,Molecular Biology ,Peptide sequence ,Alanine ,chemistry.chemical_classification ,Binding Sites ,General Medicine ,Recombinant Proteins ,Rats ,Kinetics ,Amino Acid Substitution ,Biochemistry ,chemistry ,Secretin receptor ,Pharmacophore - Abstract
The amino terminus of the secretin receptor (SecR) is known to be critical for natural agonist action, although the role it plays is still unclear. We have demonstrated that photolabile residues within both the amino-terminal (position 6) and carboxyl-terminal (positions 22 and 26) halves of secretin each covalently label receptor amino-terminal tail residues [Dong et al., J Biol Chem, 274:19161-19167 (1999), 274:903-909 (1999), and 275:26032-26039 (2000)]. Here, we extend this series of studies with an additional probe having its site of covalent attachment in a distinct region of the peptide, between amino- and carboxyl-terminal helical domains. This probe incorporated a photolabile (epsilon-p-benzoylbenzoyl)lysine in position 18 and a site for oxidative radioiodination [(tyrosine(10),(benzoyl-benzoyl)lysine(18))rat secretin-27]. This analog represented a full agonist, stimulating cAMP accumulation in Chinese hamster ovary-SecR cells in a concentration-dependent manner. It bound to the SecR specifically and saturably, and was able to efficiently label that molecule within its amino terminus. Sequential specific cleavage, purification, and sequencing demonstrated that this probe labeled receptor residue arginine(14), in the same subdomain as that labeled by previous probes. Consistent with the importance of this residue, alanine replacement mutagenesis (R14A) resulted in substantial reductions in the potency (127-fold) and binding affinity (400-fold) of secretin relative to its action at the wild-type receptor. We have been able to accommodate all four extant pairs of residue-residue approximations between divergent regions of the secretin pharmacophore and the first forty residues of the SecR into a credible molecular model of this interaction. Additional experimentally derived constraints will be necessary to determine the spatial positioning of this complex with the remainder of the SecR.
- Published
- 2002
27. Interaction between Active Pak1 and Raf-1 Is Necessary for Phosphorylation and Activation of Raf-1
- Author
-
Cynthia Hayne, Mengwei Zang, and Zhijun Luo
- Subjects
Mutant ,Plasma protein binding ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Cell Line ,chemistry.chemical_compound ,PAK1 ,Animals ,Humans ,Phosphorylation ,Binding site ,p21-activated kinases ,Molecular Biology ,DNA Primers ,Binding Sites ,Base Sequence ,Nocodazole ,Wild type ,Cell Biology ,Proto-Oncogene Proteins c-raf ,p21-Activated Kinases ,chemistry ,Tetradecanoylphorbol Acetate ,Protein Binding - Abstract
Activation of Raf-1 is a complex process in which phosphorylation of Ser(338)-Tyr(341) is a critical step. Previous studies have shown that Pak1/2 is implicated in both Ras-dependent and -independent activation of Raf-1 by phosphorylating Raf Ser(338). The present study explores the structural basis of Raf-1 phosphorylation by Pak1. We found that Pak directly associates with Raf-1 under both physiological and overexpressed conditions. The association is greatly stimulated by 4beta-12-O-tetradecanoylphorbol-13-acetate and nocodazole and by expression of the active mutants of Rac and Ras. The active forms of Pak generated by mutation of Thr(423) to Glu or truncation of the amino-terminal moiety exhibit a greater binding to Raf than the wild type, whereas the kinase-dead mutant Pak barely binds Raf. The extent of binding to Raf-1 is correlated with the ability of Pak to phosphorylate Raf and induce mitogen-activated protein kinase activation. Furthermore, the Raf-1 binding site is defined to the carboxyl terminus of the Pak catalytic domain. In addition, our results suggest that the amino-terminal regulatory region of Raf inhibits the interaction. Taken together, the results indicate that the interaction depends on the active conformations of Pak and Raf. They also argue that Pak1 is a physiological candidate for phosphorylation of Raf Ser(338) during the course of Raf activation.
- Published
- 2002
28. Hepatic SIRT1 Attenuates Hepatic Steatosis and Controls Energy Balance in Mice by Inducing Fibroblast Growth Factor 21
- Author
-
Andrew C. Adams, Amber J. Giles, Kimberly Wong, Alexei Kharitonenkov, Qin Yang, Jianwei Jiang, Leonard Guarente, Bin Gao, Jong Woo Lee, Yu Li, Mengwei Zang, Massachusetts Institute of Technology. Department of Biology, Paul F. Glenn Center for Biology of Aging Research (Massachusetts Institute of Technology), and Guarente, Leonard Pershing
- Subjects
medicine.medical_specialty ,FGF21 ,Immunoblotting ,White adipose tissue ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,chemistry.chemical_compound ,Mice ,Sirtuin 1 ,Internal medicine ,Ketogenesis ,medicine ,Animals ,Humans ,Obesity ,Mice, Knockout ,Hepatology ,Fatty acid metabolism ,Gene Expression Profiling ,Fatty liver ,Gastroenterology ,Calorimetry, Indirect ,Fasting ,Hep G2 Cells ,medicine.disease ,Up-Regulation ,Fatty Liver ,Fibroblast Growth Factors ,Fatty acid synthase ,Endocrinology ,chemistry ,Liver ,Lipogenesis ,biology.protein ,Steatosis ,Energy Metabolism ,Biomarkers - Abstract
Background & Aims The hepatocyte-derived hormone fibroblast growth factor 21 (FGF21) is a hormone-like regulator of metabolism. The nicotinamide adenine dinucleotide–dependent deacetylase SIRT1 regulates fatty acid metabolism through multiple nutrient sensors. Hepatic overexpression of SIRT1 reduces steatosis and glucose intolerance in obese mice. We investigated mechanisms by which SIRT1 controls hepatic steatosis in mice. Methods Liver-specific SIRT1 knockout (SIRT1 LKO) mice and their wild-type littermates (controls) were divided into groups that were placed on a normal chow diet, fasted for 24 hours, or fasted for 24 hours and then fed for 6 hours. Liver tissues were collected and analyzed by histologic examination, gene expression profiling, and real-time polymerase chain reaction assays. Human HepG2 cells were incubated with pharmacologic activators of SIRT1 (resveratrol or SRT1720) and mitochondrion oxidation consumption rate and immunoblot analyses were performed. FGF21 was overexpressed in SIRT1 LKO mice using an adenoviral vector. Energy expenditure was assessed by indirect calorimetry. Results Prolonged fasting induced lipid deposition in livers of control mice, but severe hepatic steatosis in SIRT1 LKO mice. Gene expression analysis showed that fasting up-regulated FGF21 in livers of control mice but not in SIRT1 LKO mice. Decreased hepatic and circulating levels of FGF21 in fasted SIRT1 LKO mice were associated with reduced hepatic expression of genes involved in fatty acid oxidation and ketogenesis, and increased expression of genes that control lipogenesis, compared with fasted control mice. Resveratrol or SRT1720 each increased the transcriptional activity of the FGF21 promoter (–2070/+117) and levels of FGF21 messenger RNA and protein in HepG2 cells. Surprisingly, SIRT1 LKO mice developed late-onset obesity with impaired whole-body energy expenditure. Hepatic overexpression of FGF21 in SIRT1 LKO mice increased the expression of genes that regulate fatty acid oxidation, decreased fasting-induced steatosis, reduced obesity, increased energy expenditure, and promoted browning of white adipose tissue. Conclusions SIRT1-mediated activation of FGF21 prevents liver steatosis caused by fasting. This hepatocyte-derived endocrine signaling appears to regulate expression of genes that control a brown fat-like program in white adipose tissue, energy expenditure, and adiposity. Strategies to activate SIRT1 or FGF21 could be used to treat fatty liver disease and obesity.
- Published
- 2013
29. The Dysregulation of AMPK Suppresses Phosphorylation of Sterol Regulatory Element Binding Protein and Increases Its Activity in the Development of Atherosclerosis in Pig and Human Diabetes
- Author
-
Mengwei Zang, Yu Li, Richard A. Cohen, Shanqin Xu, and Bingbing Jiang
- Subjects
Chemistry ,Diabetes mellitus ,Genetics ,medicine ,AMPK ,Phosphorylation ,medicine.disease ,Molecular Biology ,Biochemistry ,Biotechnology ,Sterol regulatory element-binding protein ,Cell biology - Published
- 2013
30. Retinoic Acid Receptor β Stimulates Hepatic Induction of Fibroblast Growth Factor 21 to Promote Fatty Acid Oxidation and Control Whole-body Energy Homeostasis in Mice*
- Author
-
Kimberly Wong, Mengwei Zang, Yu Li, Kenneth Walsh, and Bin Gao
- Subjects
Male ,medicine.medical_specialty ,FGF21 ,Receptors, Retinoic Acid ,Retinoic acid ,Peroxisome proliferator-activated receptor ,Biology ,Response Elements ,Biochemistry ,chemistry.chemical_compound ,Mice ,Internal medicine ,medicine ,Peroxisomes ,Animals ,Humans ,PPAR alpha ,Receptor ,Molecular Biology ,chemistry.chemical_classification ,Retinoic Acid Receptor alpha ,Fatty Acids ,Lipid metabolism ,Cell Biology ,Hep G2 Cells ,Lipid Metabolism ,Fibroblast Growth Factors ,Retinoic acid receptor ,Endocrinology ,Metabolism ,Nuclear receptor ,chemistry ,Liver ,Retinoic acid receptor alpha ,Energy Metabolism ,Oxidation-Reduction - Abstract
Activation of retinoic acid receptor (RAR) with all-trans-retinoic acid (RA) ameliorates glucose intolerance and insulin resistance in obese mice. The recently discovered fibroblast growth factor 21 (FGF21) is a hepatocyte-derived hormone that restores glucose and lipid homeostasis in obesity-induced diabetes. However, whether hepatic RAR is linked to FGF21 in the control of lipid metabolism and energy homeostasis remains elusive. Here we identify FGF21 as a direct target gene of RARβ. The gene transcription of Fgf21 is increased by the RAR agonist RA and by overexpression of RARα and RARβ, but it is unaffected by RARγ in HepG2 cells. Promoter deletion analysis characterizes a putative RA-responsive element (RARE) primarily located in the 5′-flanking region of the Fgf21 gene. Disruption of the RARE sequence abolishes RA responsiveness. In vivo adenoviral overexpression of RARβ in the liver enhances production and secretion of FGF21, which in turn promotes hepatic fatty acid oxidation and ketogenesis and ultimately leads to increased energy expenditure in mice. The metabolic effects of RA or RARβ are mimicked by FGF21 overexpression and largely abolished by FGF21 knockdown. Moreover, hepatic RARβ is bound to the putative RAREs of the Fgf21 promoter in a fasting-inducible manner in vivo, which contributes to FGF21 induction and the metabolic adaptation to prolonged fasting. In addition to other nuclear receptors, such as peroxisome proliferator-activated receptor α and retinoic acid receptor-related receptor α, RAR may act as a novel component to induce hepatic FGF21 in the regulation of lipid metabolism. The hepatic RAR-FGF21 pathway may represent a potential drug target for treating metabolic disorders.
- Published
- 2013
31. Activation of sterol regulatory element binding protein and NLRP3 inflammasome in atherosclerotic lesion development in diabetic pigs
- Author
-
Richard A. Cohen, Shanqin Xu, Mengwei Zang, Yu Li, and Bingbing Jiang
- Subjects
Male ,Inflammasomes ,Swine ,Interleukin-1beta ,Gene Expression ,lcsh:Medicine ,Coronary Artery Disease ,AMP-Activated Protein Kinases ,030204 cardiovascular system & hematology ,Cardiovascular ,Biochemistry ,Mice ,Endocrinology ,0302 clinical medicine ,AMP-activated protein kinase ,Molecular Cell Biology ,Serine ,Signaling in Cellular Processes ,Phosphorylation ,lcsh:Science ,Aorta ,0303 health sciences ,Multidisciplinary ,Protein Kinase Signaling Cascade ,biology ,Chemistry ,Fatty liver ,Acetylation ,Inflammasome ,Lipids ,Signaling Cascades ,Lipid Signaling ,Disease Progression ,Medicine ,lipids (amino acids, peptides, and proteins) ,Cellular Types ,medicine.symptom ,Sterol Regulatory Element Binding Protein 1 ,Signal Transduction ,Sterol Regulatory Element Binding Protein 2 ,Research Article ,medicine.drug ,Clinical Research Design ,Vascular Cell Adhesion Molecule-1 ,Inflammation ,Diabetes Complications ,03 medical and health sciences ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Humans ,Animal Models of Disease ,Protein Kinase Inhibitors ,Biology ,030304 developmental biology ,Diabetic Endocrinology ,lcsh:R ,Immunity ,Endothelial Cells ,AMPK ,Diabetes Mellitus Type 1 ,Atherosclerosis ,Lipid Metabolism ,medicine.disease ,Sterol regulatory element-binding protein ,Metabolism ,Gene Expression Regulation ,Proteolysis ,Immunology ,Cancer research ,biology.protein ,Clinical Immunology ,lcsh:Q ,Sterol regulatory element-binding protein 2 ,Carrier Proteins ,Biomarkers - Abstract
Background Aberrantly elevated sterol regulatory element binding protein (SREBP), the lipogenic transcription factor, contributes to the development of fatty liver and insulin resistance in animals. Our recent studies have discovered that AMP-activated protein kinase (AMPK) phosphorylates SREBP at Ser-327 and inhibits its activity, represses SREBP-dependent lipogenesis, and thereby ameliorates hepatic steatosis and atherosclerosis in insulin-resistant LDLR−/− mice. Chronic inflammation and activation of NLRP3 inflammasome have been implicated in atherosclerosis and fatty liver disease. However, whether SREBP is involved in vascular lipid accumulation and inflammation in atherosclerosis remains largely unknown. Principal Findings The preclinical study with aortic pouch biopsy specimens from humans with atherosclerosis and diabetes shows intense immunostaining for SREBP-1 and the inflammatory marker VCAM-1 in atherosclerotic plaques. The cleavage processing of SREBP-1 and -2 and expression of their target genes are increased in the well-established porcine model of diabetes and atherosclerosis, which develops human-like, complex atherosclerotic plaques. Immunostaining analysis indicates an elevation in SREBP-1 that is primarily localized in endothelial cells and in infiltrated macrophages within fatty streaks, fibrous caps with necrotic cores, and cholesterol crystals in advanced lesions. Moreover, concomitant suppression of NAD-dependent deacetylase SIRT1 and AMPK is observed in atherosclerotic pigs, which leads to the proteolytic activation of SREBP-1 by diminishing the deacetylation and Ser-372 phosphorylation of SREBP-1. Aberrantly elevated NLRP3 inflammasome markers are evidenced by increased expression of inflammasome components including NLPR3, ASC, and IL-1β. The increase in SREBP-1 activity and IL-1β production in lesions is associated with vascular inflammation and endothelial dysfunction in atherosclerotic pig aorta, as demonstrated by the induction of NF-κB, VCAM-1, iNOS, and COX-2, as well as by the repression of eNOS. Conclusions These translational studies provide in vivo evidence that the dysregulation of SIRT1-AMPK-SREBP and stimulation of NLRP3 inflammasome may contribute to vascular lipid deposition and inflammation in atherosclerosis.
- Published
- 2013
32. High-fat diet increases and the polyphenol, S17834, decreases acetylation of the sirtuin-1-dependent lysine-382 on p53 and apoptotic signaling in atherosclerotic lesion-prone aortic endothelium of normal mice
- Author
-
Chaomei Shi, Tony Verbeuren, Bingbing Jiang, Mengwei Zang, Markus Bachschmid, Richard A. Cohen, Shanqin Xu, and Xiuyun Hou
- Subjects
Male ,medicine.medical_specialty ,Endothelium ,Lysine ,Aorta, Thoracic ,Apoptosis ,Biology ,Diet, High-Fat ,Article ,Mice ,Sirtuin 1 ,Superoxides ,Internal medicine ,medicine ,Animals ,Benzopyrans ,Cells, Cultured ,Hypolipidemic Agents ,Pharmacology ,Mice, Knockout ,Caspase 3 ,Polyphenols ,Acetylation ,Atherosclerosis ,Lipids ,Endothelial stem cell ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,Biochemistry ,biology.protein ,Tumor necrosis factor alpha ,Endothelium, Vascular ,Tumor Suppressor Protein p53 ,Cardiology and Cardiovascular Medicine ,Lipoprotein ,Signal Transduction - Abstract
Our purpose was to determine if high-fat diet and treatment with a polyphenol regulate the acetylation of lysine-382 of p53, the site regulated by sirtuin-1, and apoptosis in the endothelium of the atherosclerotic lesion-prone mouse aortic arch. In cultured endothelial cells, 2 atherogenic stimuli, hydrogen peroxide and tumor necrosis factor-α, increased the acetylation of p53 lysine-382, and caspase-3 cleavage, an indicator of apoptotic signaling. The polyphenol, S17834, significantly prevented these changes. In low-density lipoprotein receptor-deficient mice, a high-fat diet increased, and treatment with S17834 attenuated early atherosclerotic lesions on the lesser curvature of the aortic arch. In wild-type C57BL6 mice fed the same diet, no atherosclerotic lesions were observed in this lesion-prone area, but p53 acetylation and caspase-3 cleavage increased in the endothelium. In high-fat fed mice, S17834 increased sirtuin-1 protein in the lesion-prone endothelium and prevented both the increase in p53 acetylation and caspase-3 cleavage without affecting blood lipids. These results indicate that high-fat diet increases and S17834 decreases the acetylation of p53 in lesion-prone aortic endothelial cells of normal mice independently of blood lipids, suggesting that the polyphenol may regulate endothelial cell p53 acetylation and apoptosis via local actions.
- Published
- 2011
33. Hepatic overexpression of SIRT1 in mice attenuates endoplasmic reticulum stress and insulin resistance in the liver
- Author
-
Ji Li, Yu Li, Mengwei Zang, Kenneth Walsh, Amber J. Giles, Xiuyun Hou, Shanqin Xu, Leonard Guarente, Zhijun Luo, Gizem Donmez, Jong Woo Lee, and Kazuto Nakamura
- Subjects
Male ,medicine.medical_specialty ,endocrine system diseases ,Immunoblotting ,Mice, Obese ,mTORC1 ,Biology ,Endoplasmic Reticulum ,Biochemistry ,Research Communications ,Mice ,Insulin resistance ,Sirtuin 1 ,Internal medicine ,Genetics ,medicine ,Glucose homeostasis ,Animals ,Humans ,Obesity ,Molecular Biology ,Endoplasmic Reticulum Chaperone BiP ,Cells, Cultured ,Mice, Knockout ,Glucose tolerance test ,medicine.diagnostic_test ,Reverse Transcriptase Polymerase Chain Reaction ,Endoplasmic reticulum ,Hep G2 Cells ,Glucose Tolerance Test ,medicine.disease ,Immunohistochemistry ,Mice, Inbred C57BL ,Insulin receptor ,Endocrinology ,Liver ,Receptors, LDL ,Unfolded protein response ,biology.protein ,Unfolded Protein Response ,biological phenomena, cell phenomena, and immunity ,Insulin Resistance ,Biotechnology - Abstract
Endoplasmic reticulum (ER) stress has been implicated in the pathophysiology of human type 2 diabetes (T2DM). Although SIRT1 has a therapeutic effect on metabolic deterioration in T2DM, the precise mechanisms by which SIRT1 improves insulin resistance remain unclear. Here, we demonstrate that adenovirus-mediated overexpression of SIRT1 in the liver of diet-induced insulin-resistant low-density lipoprotein receptor-deficient mice and of genetically obese ob/ob mice attenuates hepatic steatosis and ameliorates systemic insulin resistance. These beneficial effects were associated with decreased mammalian target of rapamycin complex 1 (mTORC1) activity, inhibited the unfolded protein response (UPR), and enhanced insulin receptor signaling in the liver, leading to decreased hepatic gluconeogenesis and improved glucose tolerance. The tunicamycin-induced splicing of X-box binding protein-1 and expression of GRP78 and CHOP were reduced by resveratrol in cultured cells in a SIRT1-dependent manner. Conversely, SIRT1-deficient mouse embryonic fibroblasts challenged with tunicamycin exhibited markedly increased mTORC1 activity and impaired ER homeostasi and insulin signaling. These effects were abolished by mTORC1 inhibition by rapamycin in human HepG2 cells. These studies indicate that SIRT1 serves as a negative regulator of UPR signaling in T2DM and that SIRT1 attenuates hepatic steatosis, ameliorates insulin resistance, and restores glucose homeostasis, largely through the inhibition of mTORC1 and ER stress.—Li, Y., Xu, S., Giles, A., Nakamura, K., Lee, J. W., Hou, X., Donmez, G., Li, J., Luo, Z., Walsh, K., Guarente, L., Zang, M. Hepatic overexpression of SIRT1 in mice attenuates endoplasmic reticulum stress and insulin resistance in the liver.
- Published
- 2011
34. AMPK exerts dual regulatory effects on the PI3K pathway
- Author
-
Xixi Luo, Mengwei Zang, Rong Tao, Jun Gong, Zhijun Luo, Wen Guo, and Rong Wen
- Subjects
0303 health sciences ,medicine.medical_specialty ,biology ,AMPK ,P70-S6 Kinase 1 ,Cell Biology ,Biochemistry ,IRS1 ,03 medical and health sciences ,Insulin receptor ,0302 clinical medicine ,Endocrinology ,030220 oncology & carcinogenesis ,Internal medicine ,Research article ,biology.protein ,medicine ,PTEN ,Phosphorylation ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,030304 developmental biology - Abstract
Background: AMP-activated protein kinase (AMPK) is a fuel-sensing enzyme that is activated when cells experience energy deficiency and conversely suppressed in surfeit of energy supply. AMPK activation improves insulin sensitivity via multiple mechanisms, among which AMPK suppresses mTOR/S6K-mediated negative feedback regulation of insulin signaling.Results: In the present study we further investigated the mechanism of AMPK-regulated insulin signaling. Our results showed that 5-aminoimidazole-4-carboxamide-1 ribonucleoside (AICAR) greatly enhanced the ability of insulin to stimulate the insulin receptor substrate-1 (IRS1)-associated PI3K activity in differentiated 3T3-F442a adipocytes, leading to increased Akt phosphorylation at S473, whereas insulin-stimulated activation of mTOR was diminished. In 3T3-F442a preadipocytes, these effects were attenuated by expression of a dominant negative mutant of AMPK a1 subunit. The enhancing effect of ACIAR on Akt phosphorylation was also observed when the cells were treated with EGF, suggesting that it is regulated at a step beyond IR/IRS1. Indeed, when the cells were chronically treated with AICAR in the absence of insulin, Akt phosphorylation was progressively increased. This event was associated with an increase in levels of phosphatidylinositol -3,4,5-trisphosphate (PIP3) and blocked by Wortmannin. We then expressed the dominant negative mutant of PTEN (C124S) and found that the inhibition of endogenous PTEN per se did not affect phosphorylation of Akt at basal levels or upon treatment with AICAR or insulin. Thus, this result suggests that AMPK activation of Akt is not mediated by regulating phosphatase and tensin homologue (PTEN).Conclusion: Our present study demonstrates that AMPK exerts dual effects on the PI3K pathway, stimulating PI3K/Akt and inhibiting mTOR/S6K.
- Published
- 2009
35. Characterization of Ser338 Phosphorylation for Raf-1 Activation*
- Author
-
Yihong Peng, Jun Gong, Martin Olbrot, Xiaoqin Xiang, Lingqi Luo, Changyan Chen, Jing Zhou, Xixi Luo, Mengwei Zang, Wei Huang, Zhijun Luo, and Qiren Huang
- Subjects
Epidermal Growth Factor ,Mutant ,Mechanisms of Signal Transduction ,Wild type ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,Phosphorylation cascade ,Cell Line ,Enzyme Activation ,Proto-Oncogene Proteins c-raf ,chemistry.chemical_compound ,Phosphoserine ,PAK1 ,chemistry ,p21-Activated Kinases ,Mutation ,Phosphorylation ,Humans ,Protein phosphorylation ,Protein Multimerization ,Molecular Biology - Abstract
Raf kinases are essential for regulating cell proliferation, survival, and tumorigenesis. However, the mechanisms by which Raf is activated are still incompletely understood. Phosphorylation plays a critical role in Raf activation in response to mitogens. The present study characterizes phosphorylation of Ser338, a crucial event for Raf-1 activation. Here we report that mutation of Lys375 to Met diminishes phosphorylation of Ser338 on both wild type Raf-1 in cells treated with epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA) and a constitutively active mutant in which Tyr340/Tyr341 are replaced by 2 aspartic acids, a conserved substitution present in natural B-Raf. The loss of Ser338 phosphorylation in these Raf mutants is not engendered by a mutation-induced conformational change, inasmuch as mutation of another site (Ser471 to Ala) in the activation segment also abolishes Ser338 phosphorylation, whereas both the kinase-dead mutants of Raf-1 are phosphorylated well by active Pak1. Furthermore, our data demonstrate that EGF-stimulated phosphorylation of Ser338 is inhibited by Sorafenib, a Raf kinase inhibitor, but not by the MEK inhibitor U0126. Interestingly, a kinase-dead mutation and Sorafenib also markedly reduce phosphorylation of Ser445 on B-Raf, a site equivalent to Raf-1 Ser338. Finally, our data reveal that Ser338 is phosphorylated on inactive Raf-1 by an active mutant of Raf-1 when they are dimerized in cells and that artificial dimerization of Raf-1 causes Ser338 phosphorylation, accompanied by activation of ERK1/2. Altogether, our data suggest that Ser338 on Raf-1 is autophosphorylated in response to mitogens.
- Published
- 2008
36. SIRT1 regulates hepatocyte lipid metabolism through activating AMP-activated protein kinase
- Author
-
Richard A. Cohen, Shanqin Xu, Yasuo Ido, Kenneth Walsh, Fan Lan, Karlene A. Maitland-Toolan, Mengwei Zang, Michel Wierzbicki, Kaori Sato, Xiuyun Hou, Bingbing Jiang, and Tony Verbeuren
- Subjects
endocrine system diseases ,Calcium-Calmodulin-Dependent Protein Kinase Kinase ,Lipids and Lipoproteins: Metabolism, Regulation, and Signaling ,Resveratrol ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Cell Line ,chemistry.chemical_compound ,Mice ,AMP-activated protein kinase ,AMP-Activated Protein Kinase Kinases ,Phenols ,Sirtuin 1 ,Multienzyme Complexes ,Animals ,Humans ,Sirtuins ,Molecular Biology ,Flavonoids ,biology ,Chemistry ,AMPK ,food and beverages ,Polyphenols ,Lipid metabolism ,Cell Biology ,Lipid Metabolism ,Cell biology ,Enzyme Activation ,Fatty acid synthase ,enzymes and coenzymes (carbohydrates) ,Glucose ,Focal Adhesion Protein-Tyrosine Kinases ,biology.protein ,Hepatocytes ,Phosphorylation ,lipids (amino acids, peptides, and proteins) ,RNA Interference ,hormones, hormone substitutes, and hormone antagonists ,Deacetylase activity ,Signal Transduction - Abstract
Resveratrol may protect against metabolic disease through activating SIRT1 deacetylase. Because we have recently defined AMPK activation as a key mechanism for the beneficial effects of polyphenols on hepatic lipid accumulation, hyperlipidemia, and atherosclerosis in type 1 diabetic mice, we hypothesize that polyphenol-activated SIRT1 acts upstream of AMPK signaling and hepatocellular lipid metabolism. Here we show that polyphenols, including resveratrol and the synthetic polyphenol S17834, increase SIRT1 deacetylase activity, LKB1 phosphorylation at Ser428, and AMPK activity. Polyphenols substantially prevent the impairment in phosphorylation of AMPK and its downstream target, ACC (acetyl-CoA carboxylase), elevation in expression of FAS (fatty acid synthase), and lipid accumulation in human HepG2 hepatocytes exposed to high glucose. These effects of polyphenols are largely abolished by pharmacological and genetic inhibition of SIRT1, suggesting that the stimulation of AMPK and lipid-lowering effect of polyphenols depend on SIRT1 activity. Furthermore, adenoviral overexpression of SIRT1 stimulates the basal AMPK signaling in HepG2 cells and in the mouse liver. AMPK activation by SIRT1 also protects against FAS induction and lipid accumulation caused by high glucose. Moreover, LKB1, but not CaMKKβ, is required for activation of AMPK by polyphenols and SIRT1. These findings suggest that SIRT1 functions as a novel upstream regulator for LKB1/AMPK signaling and plays an essential role in the regulation of hepatocyte lipid metabolism. Targeting SIRT1/LKB1/AMPK signaling by polyphenols may have potential therapeutic implications for dyslipidemia and accelerated atherosclerosis in diabetes and age-related diseases.
- Published
- 2008
37. Spatial approximation between two residues in the mid-region of secretin and the amino terminus of its receptor. Incorporation of seven sets of such constraints into a three-dimensional model of the agonist-bound secretin receptor
- Author
-
Maoqing, Dong, Zhijun, Li, Mengwei, Zang, Delia I, Pinon, Terry P, Lybrand, and Laurence J, Miller
- Subjects
Models, Molecular ,Light ,Proline ,Protein Conformation ,Molecular Sequence Data ,CHO Cells ,Ligands ,Protein Structure, Secondary ,Receptors, G-Protein-Coupled ,Receptors, Gastrointestinal Hormone ,Secretin ,Cricetinae ,Animals ,Amino Acid Sequence ,Cyanogen Bromide ,Disulfides ,Binding Sites ,Dose-Response Relationship, Drug ,Sequence Homology, Amino Acid ,Lysine ,Metalloendopeptidases ,Valine ,Precipitin Tests ,Protein Structure, Tertiary ,Rats ,Mutation ,Electrophoresis, Polyacrylamide Gel ,Peptides ,Protein Binding - Abstract
Photoaffinity labeling of receptors by bound agonists can provide important spatial constraints for molecular modeling of activated receptor complexes. Secretin is a 27-residue peptide hormone with a diffuse pharmacophoric domain that binds to the secretin receptor, a prototypic member of the Class B family of G protein-coupled receptors. In this work, we have developed, characterized, and applied two new photolabile probes for this receptor, with sites for covalent attachment in peptide positions 12 and 14, surrounding the previously most informative site of affinity labeling of this receptor. The [Tyr10,(BzBz)Lys12]rat secretin-27 probe covalently labeled receptor residue Val6, whereas the [Tyr10,(BzBz)Lys14]rat secretin-27 probe labeled receptor residue Pro38. When combined with previous photoaffinity labeling data, there are now seven independent sets of constraints distributed throughout the peptide and receptor amino-terminal domain that can be used together to generate a new molecular model of the ligand-occupied secretin receptor. The amino-terminal domain of this receptor presented a stable platform for peptide ligand interaction, with the amino terminus of the peptide hormone extended toward the transmembrane helix domain of the receptor. This provides clear insights into the molecular basis of natural ligand binding and supplies testable hypotheses regarding the molecular basis of activation of this receptor.
- Published
- 2003
38. Spatial approximation between a photolabile residue in position 13 of secretin and the amino terminus of the secretin receptor
- Author
-
Elizabeth M. Hadac, Maoqing Dong, Terry P. Lybrand, Laurence J. Miller, Xi-Qin Ding, Zhijun Li, Mengwei Zang, and Delia I. Pinon
- Subjects
Models, Molecular ,Stereochemistry ,Molecular Sequence Data ,CHO Cells ,Photoaffinity Labels ,Secretin ,Receptors, G-Protein-Coupled ,Receptors, Gastrointestinal Hormone ,chemistry.chemical_compound ,Cricetinae ,Animals ,Amino Acid Sequence ,Pharmacology ,Affinity labeling ,Photoaffinity labeling ,Edman degradation ,Ligand (biochemistry) ,Precipitin Tests ,Protein Structure, Tertiary ,Rats ,Transmembrane domain ,Biochemistry ,chemistry ,Molecular Medicine ,Secretin receptor ,Cyanogen bromide - Abstract
The amino-terminal domain of class B G protein-coupled receptors is critically important for natural peptide agonist binding and action. The precise role it plays and the molecular basis of the interaction between ligand and this domain are not well understood. In the current work, we have developed a new probe for affinity labeling the secretin receptor through a photolabile benzoyl-phenylalanine residue in position 13. This represented a high affinity ligand (K(i) = 56 +/- 8 nM) that was a potent full agonist to stimulate cellular cAMP (EC(50) = 236 +/- 22 pM). It covalently labeled the secretin receptor saturably in a single site. This was localized to the amino-terminal domain near the first transmembrane segment using a series of chemical and enzymatic digestions. Edman degradation sequencing of radiolabeled cyanogen bromide and skatole digestion products that were attached to glass beads and further cleaved with endoproteinase Asp-N demonstrated that the labeled residue represented Val(103). This is in contrast with previous photoaffinity labeling through positions 6, 18, 22, and 26 of secretin that all labeled the distal end of the amino terminus of this receptor. Together, these five pairs of residue-residue approximations provide important constraints to better understand the molecular conformation of the agonist-bound receptor.
- Published
- 2003
39. Identification of two pairs of spatially approximated residues within the carboxyl terminus of secretin and its receptor
- Author
-
Mengwei Zang, Yan W. Asmann, Delia I. Pinon, Maoqing Dong, and Laurence J. Miller
- Subjects
Phenylalanine ,Molecular Sequence Data ,Glycine ,Secretin family ,CHO Cells ,Photoaffinity Labels ,Ligands ,Biochemistry ,Protein Structure, Secondary ,Secretin ,Receptors, G-Protein-Coupled ,Receptors, Gastrointestinal Hormone ,Structure-Activity Relationship ,Protein structure ,Cricetinae ,Animals ,Amino Acid Sequence ,Receptor ,Molecular Biology ,Peptide sequence ,Alanine ,Photoaffinity labeling ,Chemistry ,Wild type ,Cell Biology ,Molecular Weight ,Kinetics ,Secretin receptor ,hormones, hormone substitutes, and hormone antagonists - Abstract
The carboxyl-terminal domains of secretin family peptides have been shown to contain key determinants for high affinity binding to their receptors. In this work, we have examined the interaction between carboxyl-terminal residues within secretin and the prototypic secretin receptor. We previously utilized photoaffinity labeling to demonstrate spatial approximation between secretin residue 22 and the receptor domain that includes the first 30 residues of the amino terminus (Dong, M., Wang, Y., Pinon, D. I., Hadac, E. M., and Miller, L. J. (1999) J. Biol. Chem. 274, 903-909). Here, we further refined the site of labeling with the p-benzoyl-phenylalanine (Bpa(22)) probe to receptor residue Leu(17) using progressive cleavage of wild type and mutant secretin receptors (V13M and V16M) and sequence analysis. We also developed a new probe incorporating a photolabile Bpa at position 26 of secretin, closer to its carboxyl terminus. This analogue was also a potent agonist (EC(50) = 72 +/- 6 pm) and bound to the secretin receptor specifically and with high affinity (K(i) = 10.3 +/- 2.4 nm). It covalently labeled the secretin receptor at a single site saturably and specifically. This was localized to the segment between residues Gly(34) and Ala(41) using chemical and enzymatic cleavage of labeled wild type and A41M mutant receptor constructs and immunoprecipitation of epitope-tagged receptor fragments. Radiochemical sequencing identified the site of covalent attachment as residue Leu(36). These new insights, along with our recent report of contact between residue 6 within the amino-terminal half of secretin and this same amino-terminal region of this receptor (Dong, M., Wang, Y., Hadac, E. M., Pinon, D. I., Holicky, E. L., and Miller, L. J. (1999) J. Biol. Chem. 274, 19161-19167), support a key role for this region, making the molecular details of this interaction of major interest.
- Published
- 2000
40. Correction to 'Spatial approximation between a photolabile residue in position 13 of secretin and the amino terminus of the secretin receptor'
- Author
-
Xi-Qin Ding, Terry P. Lybrand, Mengwei Zang, Delia I. Pinon, Laurence J. Miller, Zhijun Li, Maoqing Dong, and Elizabeth M. Hadac
- Subjects
Pharmacology ,Residue (chemistry) ,Biochemistry ,Stereochemistry ,Chemistry ,Molecular Medicine ,Secretin receptor ,Molecular Pharmacology ,Secretin - Published
- 2003
41. Molecular approximation between secretin residue 18 and the amine terminus of the secretin receptor
- Author
-
Delia I. Pinon, Maoqing Dong, Mengwei Zang, and Laurence J. Miller
- Subjects
Residue (chemistry) ,Hepatology ,Chemistry ,Stereochemistry ,Gastroenterology ,Secretin receptor ,Amine gas treating ,Secretin - Published
- 2001
42. AMPK Phosphorylates and Inhibits SREBP Activity to Attenuate Hepatic Steatosis and Atherosclerosis in Diet-Induced Insulin-Resistant Mice
- Author
-
Richard A. Cohen, Shanqin Xu, Ogyi Park, Yu Li, Mengwei Zang, Bingbing Jiang, Tony Verbeuren, Etienne Lefai, Maria M. Mihaylova, John Y.-J. Shyy, Michel Wierzbicki, Zhijun Luo, Bin Gao, Reuben J. Shaw, Bin Zheng, Xiuyun Hou, Boston University [Boston] (BU), Howard Hughes Med Inst, The Salk Institute for Biological Studies, Salk Institute for Biological Studies, Plant Molecular and Cellular Biology Laboratory, Columbia University [New York], National Institute on Alcohol Abuse and Alcoholism, Cardiovasculaire, métabolisme, diabétologie et nutrition (CarMeN), Hospices Civils de Lyon (HCL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National des Sciences Appliquées de Lyon (INSA Lyon), Institut National des Sciences Appliquées (INSA)-Université de Lyon-Institut National des Sciences Appliquées (INSA)-Université de Lyon-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Institut National de la Recherche Agronomique (INRA), University of California [San Diego] (UC San Diego), University of California, SERVIER, National Institutes of Health [RO1 DK076942, PO1 HL068758, RO1 DK080425, DK59637, R01 DK080425], and American Diabetes Association [1-08-JF-47]
- Subjects
Male ,Transcription, Genetic ,Physiology ,[SDV]Life Sciences [q-bio] ,AMP-Activated Protein Kinases ,DEFICIENT MICE ,Mice ,0302 clinical medicine ,AMP-activated protein kinase ,polycyclic compounds ,TRANSCRIPTION ,Phosphorylation ,RESVERATROL ,0303 health sciences ,biology ,ACTIVATED PROTEIN-KINASE ,FATTY LIVER ,food and beverages ,Metformin ,3. Good health ,030220 oncology & carcinogenesis ,Lipogenesis ,lipids (amino acids, peptides, and proteins) ,Sterol Regulatory Element Binding Protein 1 ,Sterol Regulatory Element Binding Protein 2 ,medicine.medical_specialty ,endocrine system ,digestive system ,Article ,LDL RECEPTOR ,03 medical and health sciences ,Insulin resistance ,LIPID-METABOLISM ,Internal medicine ,medicine ,Animals ,Humans ,Benzopyrans ,Molecular Biology ,030304 developmental biology ,AMPK ,Cell Biology ,Atherosclerosis ,medicine.disease ,Dietary Fats ,Sterol regulatory element-binding protein ,Disease Models, Animal ,ELEMENT-BINDING PROTEIN-1 ,Endocrinology ,Receptors, LDL ,CELLS ,biology.protein ,OVEREXPRESSION ,Sterol regulatory element-binding protein 2 ,Insulin Resistance ,Steatosis - Abstract
International audience; AMPK has emerged as a critical mechanism for salutary effects of polyphenols on lipid metabolic disorders in type 1 and type 2 diabetes. Here we demonstrate that AMPK interacts with and directly phosphorylates sterol regulatory element binding proteins (SREBP-1c and -2). Ser372 phosphorylation of SREBP-1c by AMPK is necessary for inhibition of proteolytic processing and transcriptional activity of SREBP-1c in response to polyphenols and metformin. AMPK stimulates Ser372 phosphorylation, suppresses SREBP-1c cleavage and nuclear translocation, and represses SREBP-1c target gene expression in hepatocytes exposed to high glucose, leading to reduced lipogenesis and lipid accumulation. Hepatic activation of AMPK by the synthetic polyphenol S17834 protects against hepatic steatosis, hyperlipidemia, and accelerated atherosclerosis in diet-induced insulin-resistant LDL receptor-deficient mice in part through phosphorylation of SREBP-1c Ser372 and suppression of SREBP-1c- and -2-dependent lipogenesis. AMPK-dependent phosphorylation of SREBP may offer therapeutic strategies to combat insulin resistance, dyslipidemia, and atherosclerosis.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.