1
|
Yeon KY, Ji S, Cheon HG. Role of activating transcription factor 3 as a mediator of the protective effects of berberine against lipopolysaccharide-stimulated SW982 cells and in rheumatoid arthritis animal models. Toxicol Appl Pharmacol 2025; 497:117279. [PMID: 40010574 DOI: 10.1016/j.taap.2025.117279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
This study aimed to explore the protective effects of berberine against rheumatoid arthritis (RA) and clarify the role of activating transcription factor 3 (ATF3) in the mechanism of action of berberine, using a lipopolysaccharide (LPS)-stimulated SW982 human synovial cell line. Berberine treatment resulted in a concentration-dependent reduction in LPS-induced proinflammatory cytokines and matrix metalloproteinases (MMPs) in SW982 cells. These inhibitory effects were associated with increased ATF3 expression, reduced nuclear translocation of nuclear factor-κB (NF-κB), and diminished phosphorylation of mitogen-activated protein kinase (MAPK). In contrast, ATF3 knockdown reversed the suppressive effects of berberine on proinflammatory cytokines and MMP production, leading to enhanced MAPK phosphorylation; however, it had minimal impact on adenosine monophosphate-activated protein kinase (AMPK) phosphorylation. Furthermore, AMPK knockdown negated the protective effects of berberine and reduced ATF3 levels, whereas treatment with 5-aminoimidazole-4-carboxamide ribonucleotide, an AMPK activator, replicated the beneficial effects of berberine. In an in vivo collagen-induced arthritis (CIA) mouse model, intraperitoneal administration of berberine significantly reduced paw edema and arthritis severity, accompanied by ATF3 induction and increased AMPK phosphorylation in the synovial tissue. These findings highlighted the pivotal role of ATF3 in mediating the protective effects of berberine in RA- and LPS-activated synoviocytes, suggesting its potential as a therapeutic agent for RA management.
Collapse
Affiliation(s)
- Kwan Yong Yeon
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon 406-799, Republic of Korea
| | - Seongmi Ji
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 406-799, Republic of Korea
| | - Hyae Gyeong Cheon
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon 406-799, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon 406-799, Republic of Korea.
| |
Collapse
|
2
|
Nicholas DA, Mbongue JC, Garcia-Pérez D, Sorensen D, Ferguson Bennit H, De Leon M, Langridge WHR. Exploring the Interplay between Fatty Acids, Inflammation, and Type 2 Diabetes. IMMUNO 2024; 4:91-107. [PMID: 39606781 PMCID: PMC11600342 DOI: 10.3390/immuno4010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Around 285 million people worldwide currently have type 2 diabetes and it is projected that this number will be surpassed by 2030. Therefore, it is of the utmost importance to enhance our comprehension of the disease's development. The regulation of diet, obesity, and inflammation in type 2 diabetes is believed to play a crucial role in enhancing insulin sensitivity and reducing the risk of onset diabetes. Obesity leads to an increase in visceral adipose tissue, which is a prominent site of inflammation in type 2 diabetes. Dyslipidemia, on the other hand, plays a significant role in attracting activated immune cells such as macrophages, dendritic cells, T cells, NK cells, and B cells to visceral adipose tissue. These immune cells are a primary source of pro-inflammatory cytokines that are believed to promote insulin resistance. This review delves into the influence of elevated dietary free saturated fatty acids and examines the cellular and molecular factors associated with insulin resistance in the initiation of inflammation induced by obesity. Furthermore, it explores novel concepts related to diet-induced inflammation and its relationship with type 2 diabetes.
Collapse
Affiliation(s)
- Dequina A. Nicholas
- School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Jacques C. Mbongue
- Department of Biological Sciences, School of Arts and Sciences, Oakwood University, Huntsville, AL 35896, USA
| | - Darysbel Garcia-Pérez
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
- Division of Molecular Genetics and Microbiology, School of Medicine Alumni Hall, Loma Linda University, Rm 102, 11021 Campus Street, Loma Linda, CA 92350, USA
| | - Dane Sorensen
- Center for Perinatal Biology, Division of Physiology, Loma Linda School of Medicine, Rm A572, 11234 Anderson Street, Loma Linda, CA 92350, USA
| | - Heather Ferguson Bennit
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| | - Marino De Leon
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| | - William H. R. Langridge
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| |
Collapse
|
3
|
Liu S, Li Z, Lan S, Hao H, Baz AA, Yan X, Gao P, Chen S, Chu Y. The Dual Roles of Activating Transcription Factor 3 (ATF3) in Inflammation, Apoptosis, Ferroptosis, and Pathogen Infection Responses. Int J Mol Sci 2024; 25:824. [PMID: 38255898 PMCID: PMC10815024 DOI: 10.3390/ijms25020824] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Transcription factors are pivotal regulators in the cellular life process. Activating transcription factor 3 (ATF3), a member of the ATF/CREB (cAMP response element-binding protein) family, plays a crucial role as cells respond to various stresses and damage. As a transcription factor, ATF3 significantly influences signal transduction regulation, orchestrating a variety of signaling pathways, including apoptosis, ferroptosis, and cellular differentiation. In addition, ATF3 serves as an essential link between inflammation, oxidative stress, and immune responses. This review summarizes the recent advances in research on ATF3 activation and its role in regulating inflammatory responses, cell apoptosis, and ferroptosis while exploring the dual functions of ATF3 in these processes. Additionally, this article discusses the role of ATF3 in diseases related to pathogenic microbial infections. Our review may be helpful to better understand the role of ATF3 in cellular responses and disease progression, thus promoting advancements in clinical treatments for inflammation and oxidative stress-related diseases.
Collapse
Affiliation(s)
- Shuang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Zhangcheng Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Shimei Lan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Huafang Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Ahmed Adel Baz
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Xinmin Yan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Pengcheng Gao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Shengli Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou 730046, China
| |
Collapse
|
4
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
5
|
Engin A. Obesity-Associated Breast Cancer: Analysis of Risk Factors and Current Clinical Evaluation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:767-819. [PMID: 39287872 DOI: 10.1007/978-3-031-63657-8_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Additionally, obese and postmenopausal women are at higher risk of all-cause and breast cancer-specific mortality compared with non-obese women with breast cancer. In this context, increased levels of estrogens, excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, adipocyte-derived adipokines, hypercholesterolemia, and excessive oxidative stress contribute to the development of breast cancer in obese women. Genetic evaluation is an integral part of diagnosis and treatment for patients with breast cancer. Despite trimodality therapy, the four-year cumulative incidence of regional recurrence is significantly higher. Axillary lymph nodes as well as primary lesions have diagnostic, prognostic, and therapeutic significance for the management of breast cancer. In clinical setting, because of the obese population primary lesions and enlarged lymph nodes could be less palpable, the diagnosis may be challenging due to misinterpretation of physical findings. Thereby, a nomogram has been created as the "Breast Imaging Reporting and Data System" (BI-RADS) to increase agreement and decision-making consistency between mammography and ultrasonography (USG) experts. Additionally, the "breast density classification system," "artificial intelligence risk scores," ligand-targeted receptor probes," "digital breast tomosynthesis," "diffusion-weighted imaging," "18F-fluoro-2-deoxy-D-glucose positron emission tomography," and "dynamic contrast-enhanced magnetic resonance imaging (MRI)" are important techniques for the earlier detection of breast cancers and to reduce false-positive results. A high concordance between estrogen receptor (ER) and progesterone receptor (PR) status evaluated in preoperative percutaneous core needle biopsy and surgical specimens is demonstrated. Breast cancer surgery has become increasingly conservative; however, mastectomy may be combined with any axillary procedures, such as sentinel lymph node biopsy (SLNB) and/or axillary lymph node dissection whenever is required. As a rule, SLNB-guided axillary dissection in breast cancer patients who have clinically axillary lymph node-positive to node-negative conversion following neoadjuvant chemotherapy is recommended, because lymphedema is the most debilitating complication after any axillary surgery. There is no clear consensus on the optimal treatment of occult breast cancer, which is much discussed today. Similarly, the current trend in metastatic breast cancer is that the main palliative treatment option is systemic therapy.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
6
|
Ke H, Chen Z, Zhao X, Yang C, Luo T, Ou W, Wang L, Liu H. Research progress on activation transcription factor 3: A promising cardioprotective molecule. Life Sci 2023:121869. [PMID: 37355225 DOI: 10.1016/j.lfs.2023.121869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
Activation transcription factor 3 (ATF3), a member of the ATF/cyclic adenosine monophosphate response element binding family, can be induced by a variety of stresses. Numerous studies have indicated that ATF3 plays multiple roles in the development and progression of cardiovascular diseases, including atherosclerosis, hypertrophy, fibrosis, myocardial ischemia-reperfusion, cardiomyopathy, and other cardiac dysfunctions. In past decades, ATF3 has been demonstrated to be detrimental to some cardiac diseases. Current studies have indicated that ATF3 can function as a cardioprotective molecule in antioxidative stress, lipid metabolic metabolism, energy metabolic regulation, and cell death modulation. To unveil the potential therapeutic role of ATF3 in cardiovascular diseases, we organized this review to explore the protective effects and mechanisms of ATF3 on cardiac dysfunction, which might provide rational evidence for the prevention and cure of cardiovascular diseases.
Collapse
Affiliation(s)
- Haoteng Ke
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zexing Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuanbin Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chaobo Yang
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Wen Ou
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lizi Wang
- Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haiqiong Liu
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
7
|
Holland SD, Ramer MS. Microglial activating transcription factor 3 upregulation: An indirect target to attenuate inflammation in the nervous system. Front Mol Neurosci 2023; 16:1150296. [PMID: 37033378 PMCID: PMC10076742 DOI: 10.3389/fnmol.2023.1150296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Activating Transcription Factor 3 (ATF3) is upregulated in reaction to several cellular stressors found in a wide range of pathological conditions to coordinate a transcriptional response. ATF3 was first implicated in the transcriptional reaction to axotomy when its massive upregulation was measured in sensory and motor neuron cell bodies following peripheral nerve injury. It has since been shown to be critical for successful axon regeneration in the peripheral nervous system and a promising target to mitigate regenerative failure in the central nervous system. However, much of the research to date has focused on ATF3's function in neurons, leaving the expression, function, and therapeutic potential of ATF3 in glia largely unexplored. In the immunology literature ATF3 is seen as a master regulator of the innate immune system. Specifically, in macrophages following pathogen or damage associated molecular pattern receptor activation and subsequent cytokine release, ATF3 upregulation abrogates the inflammatory response. Importantly, ATF3 upregulation is not exclusively due to cellular stress exposure but has been achieved by the administration of several small molecules. In the central nervous system, microglia represent the resident macrophage population and are therefore of immediate interest with respect to ATF3 induction. It is our perspective that the potential of inducing ATF3 expression to dampen inflammatory microglial phenotype represents an unexplored therapeutic target and may have synergistic benefits when paired with concomitant neuronal ATF3 upregulation. This would be of particular benefit in pathologies that involve both detrimental inflammation and neuronal damage including spinal cord injury, multiple sclerosis, and stroke.
Collapse
|
8
|
Wu J, Huang Y, Zhou X, Xiang Z, Yang Z, Meng D, Wu D, Zhang J, Yang J. ATF3 and its emerging role in atherosclerosis: a narrative review. Cardiovasc Diagn Ther 2022; 12:926-942. [PMID: 36605071 PMCID: PMC9808109 DOI: 10.21037/cdt-22-206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/08/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE Atherosclerosis (AS), is characterized by the subintima lipid accumulation and chronic inflammation inside the arterial wall, causing much mortality and morbidity worldwide. Activating transcription factor 3 (ATF3) is a member of ATF/cAMP-responsive element-binding (CREB) family of transcription factors, which acts as a master regulator of adaptive response. Recent studies have indicated the implicated role of ATF3 in atherogenesis and AS progression due to its impact on metabolic disorder, vascular injury, plaque formation, and stability. In this review, we summarize the current advances in the mechanism of ATF3 activation and the contribution of ATF3 in AS, highlighting vascular intrinsic and extrinsic mechanisms of how ATF3 influences the pathology of AS. METHODS The relevant literature (from origin to March 2022) was retrieved through PubMed research to explore the regulatory mechanism of ATF3 and the specific role of ATF3 in AS. Only English publications were reviewed in this paper. KEY CONTENT AND FINDINGS ATF3 acts as a key regulator of AS progression, which not only directly affects atherosclerotic lesions by regulating vascular homeostasis, but also gets involved in AS through systemic glucolipid metabolism and inflammatory response. The two different promoters, transcript variants, and post-translational modification in distinct cell types partly contribute to the regulatory diversity of ATF3 in AS. CONCLUSIONS ATF3 is a crucial transcription regulatory factor during atherogenesis and AS progression. Gaining a better understanding of how ATF3 affects vascular, metabolic, and immune homeostasis would advance the progress of ATF3-targeted therapy in AS.
Collapse
Affiliation(s)
- Jingyi Wu
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yifan Huang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Xiaoyan Zhou
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Zujin Xiang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Zishu Yang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Di Meng
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Di Wu
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Jing Zhang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Jian Yang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| |
Collapse
|
9
|
Chou CL, Li CH, Fang TC. Benefits of Valsartan and Amlodipine in Lipolysis through PU.1 Inhibition in Fructose-Induced Adiposity. Nutrients 2022; 14:nu14183759. [PMID: 36145135 PMCID: PMC9502698 DOI: 10.3390/nu14183759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
High fructose intake has been implicated in obesity and metabolic syndrome, which are related to increased cardiovascular mortality. However, few studies have experimentally examined the role of renin–angiotensin system blockers and calcium channel blockers (CCB) in obesity. We investigated the effects of valsartan (an angiotensin II receptor blocker) and amlodipine (a CCB) on lipolysis through the potential mechanism of PU.1 inhibition. We observed that high fructose concentrations significantly increased adipose size and triglyceride, monoacylglycerol lipase, adipose triglyceride lipase, and stearoyl-CoA desaturase-1 (SCD1), activating transcription factor 3 and PU.1 levels in adipocytes in vitro. Subsequently, PU.1 inhibitor treatment was able to reduce triglyceride, SCD1, and PU.1 levels. In addition, elevated levels of triglyceride and PU.1, stimulated by a high fructose concentration, decreased with valsartan and amlodipine treatment. Overall, these findings suggest that high fructose concentrations cause triacylglycerol storage in adipocytes through PU.1-mediated activation. Furthermore, valsartan and amlodipine treatment reduced triacylglycerol storage in adipocytes by inhibiting PU.1 activation in high fructose concentrations in vitro. Thus, the benefits of valsartan and amlodipine in lipolysis may be through PU.1 inhibition in fructose-induced adiposity, and PU.1 inhibition might have a potential therapeutic role in lipolysis in fructose-induced obesity.
Collapse
Affiliation(s)
- Chu-Lin Chou
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City 320, Taiwan
| | - Ching-Hao Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Te-Chao Fang
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: ; Tel.: +886-2-2737-2181
| |
Collapse
|
10
|
High-fat diet causes mechanical allodynia in the absence of injury or diabetic pathology. Sci Rep 2022; 12:14840. [PMID: 36050326 PMCID: PMC9437006 DOI: 10.1038/s41598-022-18281-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/09/2022] [Indexed: 12/04/2022] Open
Abstract
Understanding the interactions between diet, obesity, and diabetes is important to tease out mechanisms in painful pathology. Western diet is rich in fats, producing high amounts of circulating bioactive metabolites. However, no research has assessed how a high-fat diet (HFD) alone may sensitize an individual to non-painful stimuli in the absence of obesity or diabetic pathology. To investigate this, we tested the ability of a HFD to stimulate diet-induced hyperalgesic priming, or diet sensitization in male and female mice. Our results revealed that 8 weeks of HFD did not alter baseline pain sensitivity, but both male and female HFD-fed animals exhibited robust mechanical allodynia when exposed to a subthreshold dose of intraplantar Prostaglandin E2 (PGE2) compared to mice on chow diet. Furthermore, calcium imaging in isolated primary sensory neurons of both sexes revealed HFD induced an increased percentage of capsaicin-responsive neurons compared to their chow counterparts. Immunohistochemistry (IHC) showed a HFD-induced upregulation of ATF3, a neuronal marker of injury, in lumbar dorsal root ganglia (DRG). This suggests that a HFD induces allodynia in the absence of a pre-existing condition or injury via dietary components. With this new understanding of how a HFD can contribute to the onset of pain, we can understand the dissociation behind the comorbidities associated with obesity and diabetes to develop pharmacological interventions to treat them more efficiently.
Collapse
|
11
|
Li D, Xing Z, Yu T, Dong W, Wang Z, Peng C, Yang C. Pogostone attenuates adipose tissue inflammation by regulating the adipocyte–macrophage crosstalk via activating SIRT1. Food Funct 2022; 13:11853-11864. [DOI: 10.1039/d2fo01450e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pogostone prevents adipose tissue inflammation by activating the deacetylase SIRT1.
Collapse
Affiliation(s)
- Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziwei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tingting Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Dong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhiwei Wang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| |
Collapse
|
12
|
Hu RD, Zhang W, Li L, Zuo ZQ, Ma M, Ma JF, Yin TT, Gao CY, Yang SH, Zhao ZB, Li ZJ, Qiao GB, Lian ZX, Qu K. Chromatin accessibility analysis identifies the transcription factor ETV5 as a suppressor of adipose tissue macrophage activation in obesity. Cell Death Dis 2021; 12:1023. [PMID: 34716308 PMCID: PMC8556336 DOI: 10.1038/s41419-021-04308-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Activation of adipose tissue macrophages (ATMs) contributes to chronic inflammation and insulin resistance in obesity. However, the transcriptional regulatory machinery involved in ATM activation during the development of obesity is not fully understood. Here, we profiled the chromatin accessibility of blood monocytes and ATMs from obese and lean mice using assay for transposase-accessible chromatin sequencing (ATAC-seq). We found that monocytes and ATMs from obese and lean mice exhibited distinct chromatin accessibility status. There are distinct regulatory elements that are specifically associated with monocyte or ATM activation in obesity. We also discovered several transcription factors that may regulate monocyte and ATM activation in obese mice, specifically a predicted transcription factor named ETS translocation variant 5 (ETV5). The expression of ETV5 was significantly decreased in ATMs from obese mice and its downregulation was mediated by palmitate stimulation. The decrease in ETV5 expression resulted in macrophage activation. Our results also indicate that ETV5 suppresses endoplasmic reticulum (ER) stress and Il6 expression in macrophages. Our work delineates the changes in chromatin accessibility in monocytes and ATMs during obesity, and identifies ETV5 as a critical transcription factor suppressing ATM activation, suggesting its potential use as a therapeutic target in obesity-related chronic inflammation.
Collapse
Affiliation(s)
- Ren-Dong Hu
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wen Zhang
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Liang Li
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Zu-Qi Zuo
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Min Ma
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jin-Fen Ma
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ting-Ting Yin
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong, China
| | - Cai-Yue Gao
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shu-Han Yang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhi-Bin Zhao
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zi-Jun Li
- Guangdong Provincial Institute of Geriatrics, Concord Medical Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Gui-Bin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China.
| | - Kun Qu
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
| |
Collapse
|
13
|
Tumor necrosis factor (TNF)-α- 308 G/A gene polymorphism (rs1800629) in Egyptian patients with alopecia areata and vitiligo, a laboratory and in silico analysis. PLoS One 2020; 15:e0240221. [PMID: 33370782 PMCID: PMC7769607 DOI: 10.1371/journal.pone.0240221] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/15/2020] [Indexed: 11/19/2022] Open
Abstract
Purpose & methods Several single-nucleotide polymorphisms (SNPs) in the promoter region of the TNF-α gene can cause variations in the gene regulatory sites and act as risk factors for some autoimmune disorders as alopecia areata (AA) and vitiligo. This study aimed to detect the serum TNF-α (sTNF) level (by ELISA) and the rs1800629 (by real-time PCR) among AA and vitiligo Egyptian patients and to determine their relation with disease duration and severity. In silico analysis of this SNP to study the molecular regulation of the mutant genotypes was also done. Results In AA patients, no risk was associated with the mutant genotypes vs. the normal genotype, or with A allele vs. G allele. The risk of vitiligo was significantly higher with the G/A and A/A genotypes compared with HCs (p = 0.011). Similarly, a significantly increased risk was noted in patients with A allele vs. G allele (p<0.0001). In AA and vitiligo patients, a significant increase in sTNF-α levels was noted in the mutant G/A genotypes vs. the normal G/G genotype (p<0.0001) and in the A allele vs the G allele (p<0.0001). According to the in silico analysis, this SNP could mainly affect the SP1 transcription factor binding site with subsequent effect on TNF-α expression. Conclusion According to results of the laboratory and the in silico study, the mutant TNF-α (308) genotypes were risk factors that conferred susceptibility to vitiligo among Egyptian patients but had no effect on the susceptibility to AA.
Collapse
|
14
|
Shi Z, Zhang K, Chen T, Zhang Y, Du X, Zhao Y, Shao S, Zheng L, Han T, Hong W. Transcriptional factor ATF3 promotes liver fibrosis via activating hepatic stellate cells. Cell Death Dis 2020; 11:1066. [PMID: 33311456 PMCID: PMC7734065 DOI: 10.1038/s41419-020-03271-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
The excessive accumulation of extracellular matrix (ECM) is a key feature of liver fibrosis and the activated hepatic stellate cells (HSCs) are the major producer of ECM proteins. However, the precise mechanisms and target molecules that are involved in liver fibrosis remain unclear. In this study, we reported that activating transcription factor 3 (ATF3) was over-expressed in mice and human fibrotic livers, in activated HSCs and injured hepatocytes (HCs). Both in vivo and in vitro study have revealed that silencing ATF3 reduced the expression of pro-fibrotic genes and inhibited the activation of HSCs, thus alleviating the extent of liver fibrosis, indicating a potential protective role of ATF3 knockdown. However, ATF3 was not involved in either the apoptosis or proliferation of HCs. In addition, our data illustrated that increased nuclear localization of ATF3 promoted the transcription of fibrogenic genes and lnc-SCARNA10, which functioned as a novel positive regulator of TGF-β signaling in liver fibrogenesis by recruiting SMAD3 to the promoter of these genes. Interestingly, further study also demonstrated that lnc-SCARNA10 promoted the expression of ATF3 in a TGF-β/SMAD3-dependent manner, revealing a TGF-β/ATF3/lnc-SCARNA10 axis that contributed to liver fibrosis by activating HSCs. Taken together, our data provide a molecular mechanism implicating induced ATF3 in liver fibrosis, suggesting that ATF3 may represent a useful target in the development of therapeutic strategies for liver fibrosis.
Collapse
Affiliation(s)
- Zhemin Shi
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Kun Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yu Zhang
- Department of Hepatology and Gastroenterology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xiaoxiao Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanmian Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shuai Shao
- Department of Hepatology and Gastroenterology, Tianjin Third Central Hospital Affiliated to Nankai University, Tianjin, China
| | - Lina Zheng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tao Han
- Department of Hepatology and Gastroenterology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China. .,Department of Hepatology and Gastroenterology, Tianjin Third Central Hospital Affiliated to Nankai University, Tianjin, China. .,Tianjin Key Laboratory of Artificial Cells, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin, China.
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
15
|
Park SH, Moon Y. Enterocyte-Based Bioassay via Quantitative Combination of Proinflammatory Sentinels Specific to 8-keto-trichothecenes. Front Immunol 2020; 11:1530. [PMID: 32765531 PMCID: PMC7378738 DOI: 10.3389/fimmu.2020.01530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
Type B 8-keto-trichothecenes are muco-active mycotoxins that exist as inevitable contaminants in cereal-based foodstuffs. Gut-associated inflammation is an early frontline response during human and animal exposure to these mycotoxins. Despite various tools for chemical identification, optimized biomonitoring of sentinel response-associated biomarkers is required to assess the specific proinflammatory actions of 8-keto-trichothecenes in the gut epithelial barrier. In the present study, intoxication with 8-keto-trichothecenes in human intestinal epithelial cells was found to trigger early response gene 1 product (EGR-1) that plays crucial roles in proinflammatory chemokine induction. In contrast, epithelial exposure to 8-keto-trichothecenes resulted in downregulated expression of nuclear factor NF-kappa-B p65 protein, a key transcription factor, during general inflammatory responses in the gut. Based on the early molecular patterns of expression, the inflammation-inducing activity of 8-keto-trichothecenes was quantified using intestinal epithelial cells with dual reporters for EGR-1 and p65 proteins. EGR-1-responsive elements were linked to luciferase reporter while p65 promoter was bound to secretory alkaline phosphatase (SEAP) reporter. In response to conventional inflammagens such as endotoxins and cytokines such as TNF-α, both luciferase and SEAP activity were elevated in a dose-dependent manner. However, as expected from the mechanistic evaluation, 8-keto-trichothecene-exposed dual reporters of luciferase and SEAP displayed contrasting expression patterns. Furthermore, 8-keto-trichothecene-elevated EGR-1-responsive luciferase activity was improved by deficiency of PSMA3, an α-type subunit of the 20S proteasome core complex for ubiquitin-dependent EGR-1 degradation. This molecular event-based dual biomonitoring in epithelial cells is a promising supplementary tool for detecting typical molecular inflammatory pathways in response to 8-keto-trichothecenes in the food matrix.
Collapse
Affiliation(s)
- Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan, South Korea.,Biomedical Research Institute, Pusan National University, Yangsan, South Korea
| |
Collapse
|
16
|
Kume S, Maegawa H. Lipotoxicity, Nutrient-Sensing Signals, and Autophagy in Diabetic Nephropathy. JMA J 2020; 3:87-94. [PMID: 33150239 PMCID: PMC7590395 DOI: 10.31662/jmaj.2020-0005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetic nephropathy is a leading cause of proteinuria, kidney fibrosis, and subsequent end-stage renal disease. The renal prognosis of diabetic patients with refractory proteinuria is extremely poor. Therefore, identification of novel therapeutic targets to combat this serious condition and improve renal prognosis is urgently necessary. In diabetic patients, in addition to blood glucose levels, serum levels of free fatty acids (FFAs) are chronically elevated, even during postprandial periods. Of the various types of FFAs, saturated FFAs are highly cytotoxic and their levels are elevated in the serum of patients with diabetes. Thus, an increase in saturated FFAs is currently thought to contribute to proximal tubular cell damage and podocyte injury in diabetic nephropathy. Therefore, protecting both types of kidney cells from saturated FFA-related lipotoxicity may become a novel therapeutic approach for diabetic patients with refractory proteinuria. Interestingly, accumulating evidence suggests that controlling intracellular nutrient signals and autophagy can ameliorate the FFA-related kidney damage. Here, we review the evidence indicating possible mechanisms underlying cell injury caused by saturated FFAs and cell protective roles of intracellular nutrient signals and autophagy in diabetic nephropathy.
Collapse
Affiliation(s)
- Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
17
|
A global perspective on the crosstalk between saturated fatty acids and Toll-like receptor 4 in the etiology of inflammation and insulin resistance. Prog Lipid Res 2019; 77:101020. [PMID: 31870728 DOI: 10.1016/j.plipres.2019.101020] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022]
Abstract
Obesity is featured by chronic systemic low-grade inflammation that eventually contributes to the development of insulin resistance. Toll-like receptor 4 (TLR4) is an important mediator that triggers the innate immune response by activating inflammatory signaling cascades. Human, animal and cell culture studies identified saturated fatty acids (SFAs), the dominant non-esterified fatty acid (NEFA) in the circulation of obese subjects, as non-microbial agonists that trigger the inflammatory response via activating TLR4 signaling, which acts as an important causative link between fatty acid overload, chronic low-grade inflammation and the related metabolic aberrations. The interaction between SFAs and TLR4 may be modulated through the myeloid differentiation primary response gene 88-dependent and independent signaling pathway. Greater understanding of the crosstalk between dietary SFAs and TLR4 signaling in the pathogenesis of metabolic imbalance may facilitate the design of a more efficient pharmacological strategy to alleviate the risk of developing chronic diseases elicited in part by fatty acid overload. The current review discusses recent advances in the impact of crosstalk between SFAs and TLR4 on inflammation and insulin resistance in multiple cell types, tissues and organs in the context of metabolic dysregulation.
Collapse
|
18
|
Kumar M, Majumder D, Mal S, Chakraborty S, Gupta P, Jana K, Gupta UD, Ghosh Z, Kundu M, Basu J. Activating transcription factor 3 modulates the macrophage immune response to Mycobacterium tuberculosis infection via reciprocal regulation of inflammatory genes and lipid body formation. Cell Microbiol 2019; 22:e13142. [PMID: 31709711 DOI: 10.1111/cmi.13142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 10/20/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022]
Abstract
Infection of macrophages by Mycobacterium tuberculosis elicits an immune response that clears the bacterium. However, the bacterium is able to subvert the innate immune response. Differential expression of transcription factors (TFs) is central to the dynamic balance of this interaction. Among other functions, TFs regulate the production of antibacterial agents such as nitric oxide, pro-inflammatory cytokines and neutral lipids which are stored in lipid bodies (LBs) and favour bacterial survival. Here, we demonstrate that the TF activating transcription factor 3 (ATF3) is upregulated early during infection of macrophages or mice. Depletion of ATF3 enhances mycobacterial survival in macrophages suggesting its host-protective role. ATF3 interacts with chromatin remodelling protein brahma-related gene 1 and both associate with the promoters of interleukin-12p40, interleukin-6 and nitric oxide synthase 2, to activate expression of these genes. Strikingly, ATF3 downregulates LB formation by associating at the promoters of positive regulators of LB formation such as cholesterol 25 hydroxylase and the microRNA-33 locus. ATF3 represses the association of the activating mark, acetyl histone H4 lysine 8 at the promoter of cholesterol 25 hydroxylase. Our study suggests opposing roles of ATF3 in regulation of distinct sets of macrophage genes during infection, converging on a host-protective immune response.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Chemistry, Bose Institute, Kolkata, India
| | | | - Soumya Mal
- Department of Chemistry, Bose Institute, Kolkata, India
| | | | - Pushpa Gupta
- National JALMA Institute of Leprosy and Other Mycobacterial Disease, Agra, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Umesh D Gupta
- National JALMA Institute of Leprosy and Other Mycobacterial Disease, Agra, India
| | - Zhumur Ghosh
- Division of Bioinformatics, Bose Institute, Kolkata, India
| | | | - Joyoti Basu
- Department of Chemistry, Bose Institute, Kolkata, India
| |
Collapse
|
19
|
Cheng CF, Ku HC, Cheng JJ, Chao SW, Li HF, Lai PF, Chang CC, Don MJ, Chen HH, Lin H. Adipocyte browning and resistance to obesity in mice is induced by expression of ATF3. Commun Biol 2019; 2:389. [PMID: 31667363 PMCID: PMC6813364 DOI: 10.1038/s42003-019-0624-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 09/13/2019] [Indexed: 02/07/2023] Open
Abstract
Billions of people have obesity-related metabolic syndromes such as diabetes and hyperlipidemia. Promoting the browning of white adipose tissue has been suggested as a potential strategy, but a drug still needs to be identified. Here, genetic deletion of activating transcription factor 3 (ATF3-/- ) in mice under a high-fat diet (HFD) resulted in obesity and insulin resistance, which was abrogated by virus-mediated ATF3 restoration. ST32da, a synthetic ATF3 inducer isolated from Salvia miltiorrhiza, promoted ATF3 expression to downregulate adipokine genes and induce adipocyte browning by suppressing the carbohydrate-responsive element-binding protein-stearoyl-CoA desaturase-1 axis. Furthermore, ST32da increased white adipose tissue browning and reduced lipogenesis in HFD-induced obese mice. The anti-obesity efficacy of oral ST32da administration was similar to that of the clinical drug orlistat. Our study identified the ATF3 inducer ST32da as a promising therapeutic drug for treating diet-induced obesity and related metabolic disorders.
Collapse
MESH Headings
- 3T3-L1 Cells
- Activating Transcription Factor 3/deficiency
- Activating Transcription Factor 3/genetics
- Activating Transcription Factor 3/metabolism
- Adipocytes, Brown/metabolism
- Adipocytes, Brown/pathology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- Anti-Obesity Agents/pharmacology
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Body Temperature Regulation/physiology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Humans
- Insulin Resistance
- Lipogenesis/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/genetics
- Obesity/metabolism
- Obesity/prevention & control
- Orlistat/pharmacology
- Plant Extracts/pharmacology
- Plants, Medicinal/chemistry
- Salvia miltiorrhiza/chemistry
Collapse
Affiliation(s)
- Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Tzu Chi University, Hualien, Taiwan
| | - Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jing-Jy Cheng
- Ph.D. Program in Clinical Drug Discovery from Botanical Herbs, Taipei Medical, University, Taipei, Taiwan
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Shi-Wei Chao
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Fen Li
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Pei-Fang Lai
- Department of Emergency Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Che-Chang Chang
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jaw Don
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Hsi-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Heng Lin
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
20
|
Kim S, Song NJ, Chang SH, Bahn G, Choi Y, Rhee DK, Yun UJ, Choi J, Lee J, Yoo JH, Shin D, Park KM, Kang H, Lee S, Ku JM, Cho YS, Park KW. Sulfuretin Prevents Obesity and Metabolic Diseases in Diet Induced Obese Mice. Biomol Ther (Seoul) 2019; 27:107-116. [PMID: 30130954 PMCID: PMC6319556 DOI: 10.4062/biomolther.2018.090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/19/2018] [Accepted: 06/26/2018] [Indexed: 12/13/2022] Open
Abstract
The global obesity epidemic and associated metabolic diseases require alternative biological targets for new therapeutic strategies. In this study, we show that a phytochemical sulfuretin suppressed adipocyte differentiation of preadipocytes and administration of sulfuretin to high fat diet-fed obese mice prevented obesity and increased insulin sensitivity. These effects were associated with a suppressed expression of inflammatory markers, induced expression of adiponectin, and increased levels of phosphorylated ERK and AKT. To elucidate the molecular mechanism of sulfuretin in adipocytes, we performed microarray analysis and identified activating transcription factor 3 (Atf3) as a sulfuretin-responsive gene. Sulfuretin elevated Atf3 mRNA and protein levels in white adipose tissue and adipocytes. Consistently, deficiency of Atf3 promoted lipid accumulation and the expression of adipocyte markers. Sulfuretin’s but not resveratrol’s anti-adipogenic effects were diminished in Atf3 deficient cells, indicating that Atf3 is an essential factor in the effects of sulfuretin. These results highlight the usefulness of sulfuretin as a new anti-obesity intervention for the prevention of obesity and its associated metabolic diseases.
Collapse
Affiliation(s)
- Suji Kim
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - No-Joon Song
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seo-Hyuk Chang
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gahee Bahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yuri Choi
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ui Jeong Yun
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jinhee Choi
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jeon Lee
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyuk Yoo
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Donghan Shin
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Ki-Moon Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hee Kang
- Department of Oriental Medical Science, Graduate School of East-West Medicine, Kyunghee University, Yongin 17104, Republic of Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jin-Mo Ku
- Biomaterials Research and Development Team, Bio-Center, Gyeonggido Business Science Accelerator, Suwon 16229, Republic of Korea
| | - Yoon Shin Cho
- Department of Biomedical Science, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kye Won Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
21
|
Atf3 induction is a therapeutic target for obesity and metabolic diseases. Biochem Biophys Res Commun 2018; 504:903-908. [PMID: 30224057 DOI: 10.1016/j.bbrc.2018.09.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 09/08/2018] [Indexed: 12/17/2022]
Abstract
Activating transcription factor 3 (Atf3) has been previously demonstrated to impact obesity and metabolism. However, a metabolic role of Atf3 in mice remains debatable. We investigated the role of Atf3 in mice and further investigated Atf3 expression as a therapeutic target for obesity and metabolic diseases. Atf3 knockout (KO) mice fed with a high fat diet (HFD) aggravated weight gain and impaired glucose metabolism compared to littermate control wild type (WT) mice. Atf3 KO aged mice fed with a chow diet (CD) for longer than 10 months also displayed increased body weight and fat mass compared to WT aged mice. We also assessed requirements of Atf3 in a phytochemical mediated anti-obese effect. Effect of sulfuretin, a previously known phytochemical Atf3 inducer, in counteracting weight gain and improving glucose tolerance was almost completely abolished in the absence of Atf3, indicating that Atf3 induction can be a molecular target for preventing obesity and metabolic diseases. We further identified other Atf3 small molecule inducers that exhibit inhibitory effects on lipid accumulation in adipocytes. These data highlight the role of Atf3 in obesity and further suggest the use of chemical Atf3 inducers for prevention of obesity and metabolic diseases.
Collapse
|
22
|
Miura K, Ishioka M, Iijima K. The Roles of the Gut Microbiota and Toll-like Receptors in Obesity and Nonalcoholic Fatty Liver Disease. J Obes Metab Syndr 2017; 26:86-96. [PMID: 31089501 PMCID: PMC6484897 DOI: 10.7570/jomes.2017.26.2.86] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/28/2017] [Accepted: 03/16/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is characterized by low-grade chronic inflammation and is closely associated with the cardiovascular diseases, diabetes, and nonalcoholic fatty liver disease. Emerging data demonstrate that the gut microbiota contributes to the development of obesity by regulating the innate immune system, including the Toll-like receptors (TLRs): an altered gut microbiota composition and elevated TLR ligands are observed in obese mice and humans. The changes in the gut microbiota include an increased abundance of Firmicutes phylum and a decreased abundance of Bacteroidetes phylum. The population of beneficial bacteria that function as probiotics is decreased whereas harmful bacteria that can produce lipopolysaccharide, a TLR4 ligand, are increased in the obese state. In addition, the gut permeability is increased in obesity, which allows the delivery of larger amounts of bacterial components to the liver through the portal vein. Immune cells recognize these bacterial components through TLRs and produce diverse cytokines that kill invading pathogens. However, the sustained activation of TLR signaling induces host damage due to chronic exposure to harmful cytokines, which are produced from TLR expressing cells, including monocytes/macrophages. In the obese state, the expression of TLR is increased in several organs, including the adipose tissue and the liver. At the cell level, negative regulators of TLR signaling are suppressed, leading to activation of TLR signaling. These alterations promote inflammation in many organs. Thus, the gut microbiota and TLR signaling are therapeutic targets in patients with obesity and its related diseases.
Collapse
Affiliation(s)
- Kouichi Miura
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsuaki Ishioka
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Katsunori Iijima
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
23
|
Kang W, Wang T, Hu Z, Liu F, Sun Y, Ge S. Metformin Inhibits Porphyromonas gingivalis Lipopolysaccharide-Influenced Inflammatory Response in Human Gingival Fibroblasts via Regulating Activating Transcription Factor-3 Expression. J Periodontol 2017; 88:e169-e178. [PMID: 28548885 DOI: 10.1902/jop.2017.170168] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Chronic periodontitis, one of the most prevalent oral diseases, is associated with Porphyromonas gingivalis (Pg) lipopolysaccharide (LPS) infection and has profound effects on type 2 diabetes mellitus (t2DM). Metformin, a well-known antidiabetic agent, has been reported to exert anti-inflammatory effects on various cells. This study aims to investigate the role of metformin on LPS-influenced inflammatory response in human gingival fibroblasts (HGFs). METHODS Dose-dependent additive effects of metformin on LPS-influenced HGFs were detected. Cell-counting assay was used to determine effects of metformin and LPS on viability of HGFs. Enzyme-linked immunosorbent assay and quantitative real-time polymerase chain reaction (qRT-PCR) were applied to detect levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in differently treated cells. Activating transcription factor-3 (ATF3) small interfering (si)RNA transfection was used to determine the mechanism of metformin action, and the transfection efficiency was observed by fluorescence microscope. Effects of ATF3 knockdown were determined by qRT-PCR and Western blot. RESULTS Results showed that 5 μg/mL Pg LPS and 0.1, 0.5, and 1 mM metformin exhibited no toxicity to HGFs, and metformin inhibited LPS-influenced IL-1β, IL-6, and TNF-α production in a dose-dependent manner. Metformin and LPS could synergistically facilitate ATF3 expression, and ATF3 knockdown abolished inhibitory effects of metformin on LPS-influenced inflammatory cytokine production in HGFs. CONCLUSION The present study confirms that metformin suppresses LPS-enhanced IL-6, IL-1β, and TNF-α production in HGFs via increasing ATF3 expression.
Collapse
Affiliation(s)
- Wenyan Kang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Shandong, Jinan, China.,Department of Periodontology, School of Stomatology, Shandong University
| | - Ting Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Shandong, Jinan, China.,Department of Periodontology, School of Stomatology, Shandong University
| | - Zhekai Hu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Shandong, Jinan, China
| | - Feng Liu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Shandong University
| | - Yundong Sun
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University
| | - Shaohua Ge
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Shandong, Jinan, China.,Department of Periodontology, School of Stomatology, Shandong University
| |
Collapse
|
24
|
Nascimento DSM, Potes CS, Soares ML, Ferreira AC, Malcangio M, Castro-Lopes JM, Neto FLM. Drug-Induced HSP90 Inhibition Alleviates Pain in Monoarthritic Rats and Alters the Expression of New Putative Pain Players at the DRG. Mol Neurobiol 2017; 55:3959-3975. [PMID: 28550532 DOI: 10.1007/s12035-017-0628-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/19/2017] [Indexed: 01/17/2023]
Abstract
Purinergic receptors (P2XRs) have been widely associated with pain states mostly due to their involvement in neuron-glia communication. Interestingly, we have previously shown that satellite glial cells (SGC), surrounding dorsal root ganglia (DRG) neurons, become activated and proliferate during monoarthritis (MA) in the rat. Here, we demonstrate that P2X7R expression increases in ipsilateral DRG after 1 week of disease, while P2X3R immunoreactivity decreases. We have also reported a significant induction of the activating transcriptional factor 3 (ATF3) in MA. In this study, we show that ATF3 knocked down in DRG cell cultures does not affect the expression of P2X7R, P2X3R, or glial fibrillary acidic protein (GFAP). We suggest that P2X7R negatively regulates P2X3R, which, however, is unlikely mediated by ATF3. Interestingly, we found that ATF3 knockdown in vitro induced significant decreases in the heat shock protein 90 (HSP90) expression. Thus, we evaluated in vivo the involvement of HSP90 in MA and demonstrated that the HSP90 messenger RNA levels increase in ipsilateral DRG of inflamed animals. We also show that HSP90 is mostly found in a cleaved form in this condition. Moreover, administration of a HSP90 inhibitor, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), attenuated MA-induced mechanical allodynia in the first hours. The drug also reversed the HSP90 upregulation and cleavage. 17-DMAG seemed to attenuate glial activation and neuronal sensitization (as inferred by downregulation of GFAP and P2X3R in ipsilateral DRG) which might correlate with the observed pain alleviation. Our data indicate a role of HSP90 in MA pathophysiology, but further investigation is necessary to clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Diana Sofia Marques Nascimento
- Departamento de Biomedicina-Unidade de Biologia Experimental, Centro de Investigação Médica (CIM), Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Pain Group, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Catarina Soares Potes
- Departamento de Biomedicina-Unidade de Biologia Experimental, Centro de Investigação Médica (CIM), Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Pain Group, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Miguel Luz Soares
- Departamento de Biomedicina-Unidade de Biologia Experimental, Centro de Investigação Médica (CIM), Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Pain Group, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - António Carlos Ferreira
- Departamento de Biomedicina-Unidade de Biologia Experimental, Centro de Investigação Médica (CIM), Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Pain Group, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Marzia Malcangio
- Wolfson Centre for Age Related Diseases, King's College London, London, UK
| | - José Manuel Castro-Lopes
- Departamento de Biomedicina-Unidade de Biologia Experimental, Centro de Investigação Médica (CIM), Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,Pain Group, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Fani Lourença Moreira Neto
- Departamento de Biomedicina-Unidade de Biologia Experimental, Centro de Investigação Médica (CIM), Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal. .,Pain Group, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal. .,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
25
|
Zhou J, Edgar BA, Boutros M. ATF3 acts as a rheostat to control JNK signalling during intestinal regeneration. Nat Commun 2017; 8:14289. [PMID: 28272390 PMCID: PMC5344978 DOI: 10.1038/ncomms14289] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022] Open
Abstract
Epithelial barrier function is maintained by coordination of cell proliferation and cell loss, whereas barrier dysfunction can lead to disease and organismal death. JNK signalling is a conserved stress signalling pathway activated by bacterial infection and tissue damage, often leading to apoptotic cell death and compensatory cell proliferation. Here we show that the stress inducible transcription factor ATF3 restricts JNK activity in the Drosophila midgut. ATF3 regulates JNK-dependent apoptosis and regeneration through the transcriptional regulation of the JNK antagonist, Raw. Enterocyte-specific ATF3 inactivation increases JNK activity and sensitivity to infection, a phenotype that can be rescued by Raw overexpression or JNK suppression. ATF3 depletion enhances intestinal regeneration triggered by infection, but does not compensate for the loss of enterocytes and ATF3-depleted flies succumb to infection due to intestinal barrier dysfunction. In sum, we provide a mechanism to explain how an ATF3-Raw module controls JNK signalling to maintain normal intestinal barrier function during acute infection. Stress response JNK signalling is important for cell death-induced regeneration. Here the authors show in adult Drosophila enterocytes that ATF3 regulates the expression of Raw, a JNK antagonist, to control intestinal regeneration and barrier function in response to infection.
Collapse
Affiliation(s)
- Jun Zhou
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department for Cell and Molecular Biology, Medical Faculty Mannheim, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Bruce A Edgar
- German Cancer Research Center (DKFZ)-Center for Molecular Biology Heidelberg (ZMBH) Alliance, 69120 Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department for Cell and Molecular Biology, Medical Faculty Mannheim, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
26
|
Pereira JX, Cavalcante Y, Wanzeler de Oliveira R. The role of inflammation in adipocytolytic nonsurgical esthetic procedures for body contouring. Clin Cosmet Investig Dermatol 2017; 10:57-66. [PMID: 28260937 PMCID: PMC5327851 DOI: 10.2147/ccid.s125580] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The adipocytolytic non-surgical esthetic procedures are indicated for the reduction of localized fat and are effective in reducing local adiposity through the ablation of adipocytes with fast and lasting results, besides causing local inflammation. Objective This study aimed to characterize the adipocytolytic procedures and correlate the phases of the inflammatory process with the results obtained from such procedures, in order to clarify the role of inflammation triggered by the adipocytolytic procedures and its relation with the lipolytic process, with a focus on body shaping. Methods This work is an integrative literature review that presents a total of 72 articles published between 1998 and 2015, derived from the PubMed database, in order to establish a relationship between clinical and basic science research, assuming an important role in medical practice based on evidence. Results The results show that the adipocytolytic procedures are characterized by triggering inflammation arising from the disruption of adipocytes by interfering with the lipolytic signaling pathways in both acute and chronic phases of inflammation through the direct action of proinflammatory cytokines or catecholamines. Therefore, inflammation plays an important role in modulating the lipolytic process, influencing body shaping. Conclusion The inflammatory process assists the adipolytic process in all stages of inflammation, contributing to the reduction of body contouring.
Collapse
|
27
|
Transcriptomic Analysis of THP-1 Macrophages Exposed to Lipoprotein Hydrolysis Products Generated by Lipoprotein Lipase. Lipids 2017; 52:189-205. [DOI: 10.1007/s11745-017-4238-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/02/2017] [Indexed: 11/25/2022]
|
28
|
Ghigo A, Frati G, Sciarretta S. A novel protective role for activating transcription factor 3 in the cardiac response to metabolic stress. Cardiovasc Res 2017; 113:113-114. [PMID: 28082449 DOI: 10.1093/cvr/cvw252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy
| | - Giacomo Frati
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, Latina (LT) 04100, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Località Camerelle, Pozzilli (IS) 86077, Italy
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, Latina (LT) 04100, Italy; .,Department of AngioCardioNeurology, IRCCS Neuromed, Località Camerelle, Pozzilli (IS) 86077, Italy
| |
Collapse
|
29
|
The Pathogenesis of Obesity-Associated Adipose Tissue Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:221-245. [PMID: 28585201 DOI: 10.1007/978-3-319-48382-5_9] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
The citrus flavonoid naringenin confers protection in a murine endotoxaemia model through AMPK-ATF3-dependent negative regulation of the TLR4 signalling pathway. Sci Rep 2016; 6:39735. [PMID: 28004841 PMCID: PMC5177915 DOI: 10.1038/srep39735] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
Excessive activation of the TLR4 signalling pathway is critical for inflammation-associated disorders, while negative regulators play key roles in restraining TLR4 from over-activation. Naringenin is a citrus flavonoid with remarkable anti-inflammatory activity, but the mechanisms underlying its inhibition of LPS/TLR4 signalling are less clear. This study investigated the molecular targets and therapeutic effects of naringenin in vitro and in vivo. In LPS-stimulated murine macrophages, naringenin suppressed the expression of TNF-α, IL-6, TLR4, inducible NO synthase (iNOS), cyclo-oxygenase-2 (COX2) and NADPH oxidase-2 (NOX2). Naringenin also inhibited NF-κB and mitogen-activated protein kinase (MAPK) activation. However, it did not affect the IRF3 signalling pathway or interferon production, which upregulate activating transcription factor 3 (ATF3), an inducible negative regulator of TLR4 signalling. Naringenin was demonstrated to directly increase ATF3 expression. Inhibition of AMPK and its upstream calcium-dependent signalling reduced ATF3 expression and dampened the anti-inflammatory activity of naringenin. In murine endotoxaemia models, naringenin ameliorated pro-inflammatory reactions and improved survival. Furthermore, it induced AMPK activation in lung tissues, which was required for ATF3 upregulation and the enhanced anti-inflammatory activity. Overall, this study reveals a novel mechanism of naringenin through AMPK-ATF3-dependent negative regulation of the LPS/TLR4 signalling pathway, which thereby confers protection against murine endotoxaemia.
Collapse
|
31
|
Fernández-Verdejo R, Vanwynsberghe AM, Essaghir A, Demoulin JB, Hai T, Deldicque L, Francaux M. Activating transcription factor 3 attenuates chemokine and cytokine expression in mouse skeletal muscle after exercise and facilitates molecular adaptation to endurance training. FASEB J 2016; 31:840-851. [PMID: 27856557 DOI: 10.1096/fj.201600987r] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/31/2016] [Indexed: 12/17/2022]
Abstract
Activating transcription factor (ATF)3 regulates the expression of inflammation-related genes in several tissues under pathological contexts. In skeletal muscle, atf3 expression increases after exercise, but its target genes remain unknown. We aimed to identify those genes and to determine the influence of ATF3 on muscle adaptation to training. Skeletal muscles of ATF3-knockout (ATF3-KO) and control mice were analyzed at rest, after exercise, and after training. In resting muscles, there was no difference between genotypes in enzymatic activities or fiber type. After exercise, a microarray analysis in quadriceps revealed ATF3 affects genes modulating chemotaxis and chemokine/cytokine activity. Quantitative PCR showed that the mRNA levels of chemokine C-C motif ligand (ccl)8 and chemokine C-X-C motif ligand (cxcl)13 were higher in quadriceps of ATF3-KO mice than in control mice. The same was observed for ccl9 and cxcl13 in soleus. Also in soleus, ccl2, interleukin (il)6, il1β, and cluster of differentiation (cd)68 mRNA levels increased after exercise only in ATF3-KO mice. Endurance training increased the basal mRNA level of hexokinase-2, hormone sensitive lipase, glutathione peroxidase-1, and myosin heavy chain IIa in quadriceps of control mice but not in ATF3-KO mice. In summary, ATF3 attenuates the expression of inflammation-related genes after exercise and thus facilitates molecular adaptation to training.-Fernández-Verdejo, R., Vanwynsberghe, A. M., Essaghir, A., Demoulin, J.-B., Hai, T., Deldicque, L., Francaux, M. Activating transcription factor 3 attenuates chemokine and cytokine expression in mouse skeletal muscle after exercise and facilitates molecular adaptation to endurance training.
Collapse
Affiliation(s)
| | - Aline M Vanwynsberghe
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Ahmed Essaghir
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium; and
| | | | - Tsonwin Hai
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio, USA
| | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Marc Francaux
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium;
| |
Collapse
|
32
|
Activated Transcription Factor 3 in Association with Histone Deacetylase 6 Negatively Regulates MicroRNA 199a2 Transcription by Chromatin Remodeling and Reduces Endothelin-1 Expression. Mol Cell Biol 2016; 36:2838-2854. [PMID: 27573019 DOI: 10.1128/mcb.00345-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/25/2016] [Indexed: 01/18/2023] Open
Abstract
Previous studies showed that high levels of placenta growth factor (PlGF) correlated with increased plasma levels of endothelin-1 (ET-1), a potent vasoconstrictor, in sickle cell disease (SCD). PlGF-mediated transcription of the ET-1 gene occurs by activation of hypoxia inducible factor 1α (HIF-1α) and posttranscriptionally by microRNA 199a2 (miR-199a2), which targets the 3' untranslated region (UTR) of HIF-1α mRNA. However, relatively less is known about how PlGF represses the expression of miR-199a2 located in the DNM3 opposite strand (DNM3os) transcription unit. Here, we show that PlGF induces the expression of activated transcription factor 3 (ATF3), which, in association with accessory proteins (c-Jun dimerization protein 2 [JDP2], ATF2, and histone deacetylase 6 [HDAC6]), as determined by proteomic analysis, binds to the DNM3os promoter. Furthermore, we show that association of HDAC6 with ATF3 at its binding site in this promoter was correlated with repression of miR-199a2 transcription, as shown by DNM3os transcription reporter and chromatin immunoprecipitation (ChIP) assays. Tubacin, an inhibitor of HDAC6, antagonized PlGF-mediated repression of DNM3os/pre-miR-199a2 transcription with a concomitant reduction in ET-1 levels in cultured endothelial cells. Analysis of lung tissues from Berkeley sickle (BK-SS) mice showed increased levels of ATF3 and increased expression of ET-1. Delivery of tubacin to BK-SS mice significantly attenuated plasma ET-1 and PlGF levels. Our studies demonstrated that ATF3 in conjunction with HDAC6 acts as a transcriptional repressor of the DNM3os/miR-199a2 locus.
Collapse
|
33
|
Hachiya R, Shiihashi T, Shirakawa I, Iwasaki Y, Matsumura Y, Oishi Y, Nakayama Y, Miyamoto Y, Manabe I, Ochi K, Tanaka M, Goda N, Sakai J, Suganami T, Ogawa Y. The H3K9 methyltransferase Setdb1 regulates TLR4-mediated inflammatory responses in macrophages. Sci Rep 2016; 6:28845. [PMID: 27349785 PMCID: PMC4924096 DOI: 10.1038/srep28845] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/08/2016] [Indexed: 12/24/2022] Open
Abstract
Proinflammatory cytokine production in macrophages involves multiple regulatory mechanisms, which are affected by environmental and intrinsic stress. In particular, accumulating evidence has suggested epigenetic control of macrophage differentiation and function mainly in vitro. SET domain, bifurcated 1 (Setdb1, also known as Eset) is a histone 3 lysine 9 (H3K9)-specific methyltransferase and is essential for early development of embryos. Here we demonstrate that Setdb1 in macrophages potently suppresses Toll-like receptor 4 (TLR4)-mediated expression of proinflammatory cytokines including interleukin-6 through its methyltransferase activity. As a molecular mechanism, Setdb1-deficiency decreases the basal H3K9 methylation levels and augments TLR4-mediated NF-κB recruitment on the proximal promoter region of interleukin-6, thereby accelerating interleukin-6 promoter activity. Moreover, macrophage-specific Setdb1-knockout mice exhibit higher serum interleukin-6 concentrations in response to lipopolysaccharide challenge and are more susceptible to endotoxin shock than wildtype mice. This study provides evidence that the H3K9 methyltransferase Setdb1 is a novel epigenetic regulator of proinflammatory cytokine expression in macrophages in vitro and in vivo. Our data will shed insight into the better understanding of how the immune system reacts to a variety of conditions.
Collapse
Affiliation(s)
- Rumi Hachiya
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takuya Shiihashi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.,Department of Life Science and Medical Bio-Science, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku,Tokyo, 162-8480, Japan
| | - Ibuki Shirakawa
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yorihiro Iwasaki
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yoshihiro Matsumura
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Yumiko Oishi
- Department of Cellular and Molecular Medicine, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yukiteru Nakayama
- Department of Cardiovascular Medicine, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yoshihiro Miyamoto
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-0873, Japan
| | - Ichiro Manabe
- Department of Aging Research, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Kozue Ochi
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bio-Science, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku,Tokyo, 162-8480, Japan
| | - Juro Sakai
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan.,Japan Science and Technology Agency, PRESTO, 7 Goban-cho, Chiyoda-ku, Tokyo, 102-0076, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.,Japan Agency for Medical Research and Development, AMED-CREST, 1-7-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan.,Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| |
Collapse
|
34
|
Bae YA, Cheon HG. Activating transcription factor-3 induction is involved in the anti-inflammatory action of berberine in RAW264.7 murine macrophages. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:415-24. [PMID: 27382358 PMCID: PMC4930910 DOI: 10.4196/kjpp.2016.20.4.415] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/13/2022]
Abstract
Berberine is an isoquinoline alkaloid found in Rhizoma coptidis, and elicits anti-inflammatory effects through diverse mechanisms. Based on previous reports that activating transcription factor-3 (ATF-3) acts as a negative regulator of LPS signaling, the authors investigated the possible involvement of ATF-3 in the anti-inflammatory effects of berberine. It was found berberine concentration-dependently induced the expressions of ATF-3 at the mRNA and protein levels and concomitantly suppressed the LPS-induced productions of proinflammatory cytokines (TNF-α, IL-6, and IL-1β). In addition, ATF-3 knockdown abolished the inhibitory effects of berberine on LPS-induced proinflammatory cytokine production, and prevented the berberine-induced suppression of MAPK phosphorylation, but had little effect on AMPK phosphorylation. On the other hand, the effects of berberine, that is, ATF-3 induction, proinflammatory cytokine inhibition, and MAPK inactivation, were prevented by AMPK knockdown, suggesting ATF-3 induction occurs downstream of AMPK activation. The in vivo administration of berberine to mice with LPS-induced endotoxemia increased ATF-3 expression and AMPK phosphorylation in spleen and lung tissues, and concomitantly reduced the plasma and tissue levels of proinflammatory cytokines. These results suggest berberine has an anti-inflammatory effect on macrophages and that this effect is attributable, at least in part, to pathways involving AMPK activation and ATF-3 induction.
Collapse
Affiliation(s)
- Young-An Bae
- Department of Microbiology, Gachon University School of Medicine, Incheon 21936, Korea
| | - Hyae Gyeong Cheon
- Department of Pharmacology, Gachon University School of Medicine, Incheon 21936, Korea.; Gachon Medical Research Institute, Gil Medical Center, Incheon 21565, Korea
| |
Collapse
|
35
|
Nguyen CT, Luong TT, Lee SY, Kim GL, Pyo S, Rhee DK. ATF3 provides protection fromStaphylococcus aureusandListeria monocytogenesinfections. FEMS Microbiol Lett 2016; 363:fnw062. [DOI: 10.1093/femsle/fnw062] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2016] [Indexed: 12/22/2022] Open
|
36
|
Yang CJ, Yang J, Fan ZX, Yang J. Activating transcription factor 3--an endogenous inhibitor of myocardial ischemia-reperfusion injury (Review). Mol Med Rep 2016; 13:9-12. [PMID: 26548643 DOI: 10.3892/mmr.2015.4529] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 10/06/2015] [Indexed: 11/06/2022] Open
Abstract
Coronary heart diseases, particularly acute coronary syndrome, have increased in morbidity and mortality in recent decades. Percutaneous coronary intervention, coronary artery bypass grafting and thrombolytic agents are effective strategies to rescue the infarcted myocardium. In addition to acute myocardial infarction, the resulting myocardial ischemia‑reperfusion injury (MIRI) leads to serious secondary injury of the heart. Studies have demonstrated that activating transcription factor (ATF)/cyclic adenosine monophosphate response element binding family member ATF3 had a negative regulatory role in IRI, particularly in the kidney, cerebrum and liver. The present review expounded the expression characteristics of ATF3 and its protective effects against MIRI, providing a theoretical basis for the overexpression of ATF3 in the myocardium as a promising gene-therapeutic strategy for MIRI.
Collapse
Affiliation(s)
- Chao-Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, Yichang, Hubei 443000, P.R. China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, Yichang, Hubei 443000, P.R. China
| | - Zhi-Xing Fan
- Department of Cardiology, The First College of Clinical Medical Sciences, Yichang, Hubei 443000, P.R. China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
37
|
Velloso LA, Folli F, Saad MJ. TLR4 at the Crossroads of Nutrients, Gut Microbiota, and Metabolic Inflammation. Endocr Rev 2015; 36:245-71. [PMID: 25811237 DOI: 10.1210/er.2014-1100] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity is accompanied by the activation of low-grade inflammatory activity in metabolically relevant tissues. Studies have shown that obesity-associated insulin resistance results from the inflammatory targeting and inhibition of key proteins of the insulin-signaling pathway. At least three apparently distinct mechanisms-endoplasmic reticulum stress, toll-like receptor (TLR) 4 activation, and changes in gut microbiota-have been identified as triggers of obesity-associated metabolic inflammation; thus, they are expected to represent potential targets for the treatment of obesity and its comorbidities. Here, we review the data that place TLR4 in the center of the events that connect the consumption of dietary fats with metabolic inflammation and insulin resistance. Changes in the gut microbiota can lead to reduced integrity of the intestinal barrier, leading to increased leakage of lipopolysaccharides and fatty acids, which can act upon TLR4 to activate systemic inflammation. Fatty acids can also trigger endoplasmic reticulum stress, which can be further stimulated by cross talk with active TLR4. Thus, the current data support a connection among the three main triggers of metabolic inflammation, and TLR4 emerges as a link among all of these mechanisms.
Collapse
Affiliation(s)
- Licio A Velloso
- Department of Internal Medicine (L.A.V., F.F., M.J.S.), University of Campinas, 13084-970 Campinas SP, Brazil; and Department of Medicine (F.F.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Franco Folli
- Department of Internal Medicine (L.A.V., F.F., M.J.S.), University of Campinas, 13084-970 Campinas SP, Brazil; and Department of Medicine (F.F.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Mario J Saad
- Department of Internal Medicine (L.A.V., F.F., M.J.S.), University of Campinas, 13084-970 Campinas SP, Brazil; and Department of Medicine (F.F.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
38
|
Li W, Wang L, Huang W, Skibba M, Fang Q, Xie L, Wei T, Feng Z, Liang G. Inhibition of ROS and inflammation by an imidazopyridine derivative X22 attenuate high fat diet-induced arterial injuries. Vascul Pharmacol 2015; 72:153-62. [PMID: 25989105 DOI: 10.1016/j.vph.2015.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/05/2015] [Accepted: 05/09/2015] [Indexed: 11/24/2022]
Abstract
Obesity is strongly associated with the cause of structural and functional changes of the artery. Oxidative stress and inflammation play a critical role in the development of obesity-induced cardiovascular disorders. Our group previously found that an imidazopyridine derivative X22 showed excellent anti-inflammatory activity in LPS-stimulated macrophages. This study was designed to investigate the protective effects of X22 on high fat diet (HFD)-induced arterial injury and its underlying mechanisms. We observed that palmitate (PA) treatment in HUVECs induced a marked increase in reactive oxygen species, inflammation, apoptosis, and fibrosis. All of these changes were effectively suppressed by X22 treatment in a dose-dependent manner, associated with NF-κB inactivation and Nrf-2 activation. In HFD-fed rats, administration of X22 at 10mg/kg significantly decreased the arterial inflammation and oxidative stress, and eventually improved the arterial matrix remodeling and apoptosis. X22 at 10mg/kg showed a comparable bioactivity with the positive control, curcumin at 50mg/kg. The in vivo beneficial effects of X22 are also associated with its ability to increase Nrf2 expression and inhibit NF-κB activation in the artery of HFD-fed rats. Overall, these results suggest that X22 may have therapeutic potential in the treatment of obesity-induced artery injury via regulation of Nrf2-mediated oxidative stress and NF-κB-mediated inflammation.
Collapse
Affiliation(s)
- Weixin Li
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Cardiology, the Fifth Affiliated Hospital, Wenzhou Medical University, Lishui, Zhejiang, China
| | - Lintao Wang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Melissa Skibba
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qilu Fang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Longteng Xie
- Department of Infectious Diseases, The Affiliated Xiangshan Hospital, Wenzhou Medical University, Xiangshan, Zhejiang, China
| | - Tiemin Wei
- Department of Cardiology, the Fifth Affiliated Hospital, Wenzhou Medical University, Lishui, Zhejiang, China
| | - Zhiguo Feng
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Infectious Diseases, The Affiliated Xiangshan Hospital, Wenzhou Medical University, Xiangshan, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
39
|
High-fat diet decreases energy expenditure and expression of genes controlling lipid metabolism, mitochondrial function and skeletal system development in the adipose tissue, along with increased expression of extracellular matrix remodelling- and inflammation-related genes. Br J Nutr 2015; 113:867-77. [PMID: 25744306 DOI: 10.1017/s0007114515000100] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The aim of the present study was to identify the genes differentially expressed in the visceral adipose tissue in a well-characterised mouse model of high-fat diet (HFD)-induced obesity. Male C57BL/6J mice (n 20) were fed either HFD (189 % of energy from fat) or low-fat diet (LFD, 42 % of energy from fat) for 16 weeks. HFD-fed mice exhibited obesity, insulin resistance, dyslipidaemia and adipose collagen accumulation, along with higher levels of plasma leptin, resistin and plasminogen activator inhibitor type 1, although there were no significant differences in plasma cytokine levels. Energy intake was similar in the two diet groups owing to lower food intake in the HFD group; however, energy expenditure was also lower in the HFD group than in the LFD group. Microarray analysis revealed that genes related to lipolysis, fatty acid metabolism, mitochondrial energy transduction, oxidation-reduction, insulin sensitivity and skeletal system development were down-regulated in HFD-fed mice, and genes associated with extracellular matrix (ECM) components, ECM remodelling and inflammation were up-regulated. The top ten up- or down-regulated genes include Acsm3, mt-Nd6, Fam13a, Cyp2e1, Rgs1 and Gpnmb, whose roles in the deterioration of obesity-associated adipose tissue are poorly understood. In conclusion, the genes identified here provide new therapeutic opportunities for prevention and treatment of diet-induced obesity.
Collapse
|
40
|
Huang L, Zhang SM, Zhang P, Zhang XJ, Zhu LH, Chen K, Gao L, Zhang Y, Kong XJ, Tian S, Zhang XD, Li H. Interferon regulatory factor 7 protects against vascular smooth muscle cell proliferation and neointima formation. J Am Heart Assoc 2014; 3:e001309. [PMID: 25304854 PMCID: PMC4323813 DOI: 10.1161/jaha.114.001309] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Interferon regulatory factor 7 (IRF7), a member of the interferon regulatory factor family, plays important roles in innate immunity and immune cell differentiation. However, the role of IRF7 in neointima formation is currently unknown. Methods and Results Significant decreases in IRF7 expression were observed in vascular smooth muscle cells (VSMCs) following carotid artery injury in vivo and platelet‐derived growth factor‐BB (PDGF‐BB) stimulation in vitro. Compared with non‐transgenic (NTG) controls, SMC‐specific IRF7 transgenic (IRF7‐TG) mice displayed reduced neointima formation and VSMC proliferation in response to carotid injury, whereas a global knockout of IRF7 (IRF7‐KO) resulted in the opposite effect. Notably, a novel IRF7‐KO rat strain was successfully generated and used to further confirm the effects of IRF7 deletion on the acceleration of intimal hyperplasia based on a balloon injury‐induced vascular lesion model. Mechanistically, IRF7's inhibition of carotid thickening and the expression of VSMC proliferation markers was dependent on the interaction of IRF7 with activating transcription factor 3 (ATF3) and its downstream target, proliferating cell nuclear antigen (PCNA). The evidence that IRF7/ATF3‐double‐TG (DTG) and IRF7/ATF3‐double‐KO (DKO) mice abolished the regulatory effects exhibited by the IRF7‐TG and IRF7‐KO mice, respectively, validated the underlying molecular events of IRF7‐ATF3 interaction. Conclusions These findings demonstrated that IRF7 modulated VSMC proliferation and neointima formation by interacting with ATF3, thereby inhibiting the ATF3‐mediated induction of PCNA transcription. The results of this study indicate that IRF7 is a novel modulator of neointima formation and VSMC proliferation and may represent a promising target for vascular disease therapy.
Collapse
Affiliation(s)
- Ling Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Shu-Min Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Xiao-Jing Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China (X.J.Z.)
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Ke Chen
- College of Life Sciences, Wuhan University, Wuhan, China (K.C., X.D.Z.)
| | - Lu Gao
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G.)
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Xiang-Jie Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| | - Xiao-Dong Zhang
- College of Life Sciences, Wuhan University, Wuhan, China (K.C., X.D.Z.)
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.) Cardiovascular Research Institute of Wuhan University, Wuhan, China (L.H., S.M.Z., P.Z., L.H.Z., Y.Z., X.J.K., S.T., H.L.)
| |
Collapse
|
41
|
Kračmerová J, Czudková E, Koc M, Mališová L, Šiklová M, Štich V, Rossmeislová L. Postprandial inflammation is not associated with endoplasmic reticulum stress in peripheral blood mononuclear cells from healthy lean men. Br J Nutr 2014; 112:573-82. [PMID: 24870697 DOI: 10.1017/s0007114514001093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The consumption of lipids and simple sugars induces an inflammatory response whose exact molecular trigger remains elusive. The aims of the present study were to investigate (1) whether inflammation induced by a single high-energy, high-fat meal (HFM) is associated with endoplasmic reticulum stress (ERS) in peripheral blood mononuclear cells (PBMC) and (2) whether these inflammatory and ERS responses could be prevented by the chemical chaperone ursodeoxycholic acid (UDCA). A total of ten healthy lean men were recruited to a randomised, blind, cross-over trial. Subjects were given two doses of placebo (lactose) or UDCA before the consumption of a HFM (6151 kJ; 47·4 % lipids). Blood was collected at baseline and 4 h after the HFM challenge. Cell populations and their activation were analysed using flow cytometry, and plasma levels of inflammatory cytokines were assessed by ELISA and Luminex technology. Gene expression levels of inflammatory and ERS markers were analysed in CD14⁺ and CD14⁻ PBMC using quantitative RT-PCR. The HFM induced an increase in the mRNA expression levels of pro-inflammatory cytokines (IL-1β, 2·1-fold; IL-8, 2·4-fold; TNF-α, 1·4-fold; monocyte chemoattractant protein 1, 2·1-fold) and a decrease in the expression levels of miR181 (0·8-fold) in CD14⁺ monocytes. The HFM challenge did not up-regulate the expression of ERS markers (XBP1, HSPA5, EDEM1, DNAJC3 and ATF4) in either CD14⁺ or CD14⁻ cell populations, except for ATF3 (2·3-fold). The administration of UDCA before the consumption of the HFM did not alter the HFM-induced change in the expression levels of ERS or inflammatory markers. In conclusion, HFM-induced inflammation detectable on the level of gene expression in PBMC was not associated with the concomitant increase in the expression levels of ERS markers and could not be prevented by UDCA.
Collapse
Affiliation(s)
- Jana Kračmerová
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| | - Eva Czudková
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| | - Michal Koc
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| | - Lucia Mališová
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| | - Michaela Šiklová
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| | - Vladimír Štich
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| | - Lenka Rossmeislová
- Department of Sport Medicine,Third Faculty of Medicine, Charles University in Prague, Ruská 87,100 00,Prague 10,Czech Republic
| |
Collapse
|
42
|
Park SH, Kim J, Do KH, Park J, Oh CG, Choi HJ, Song BG, Lee SJ, Kim YS, Moon Y. Activating transcription factor 3-mediated chemo-intervention with cancer chemokines in a noncanonical pathway under endoplasmic reticulum stress. J Biol Chem 2014; 289:27118-27133. [PMID: 25122760 DOI: 10.1074/jbc.m114.568717] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The cell-protective features of the endoplasmic reticulum (ER) stress response are chronically activated in vigorously growing malignant tumor cells, which provide cellular growth advantages over the adverse microenvironment including chemotherapy. As an intervention with ER stress responses in the intestinal cancer cells, preventive exposure to flavone apigenin potentiated superinduction of a regulatory transcription factor, activating transcription factor 3 (ATF3), which is also known to be an integral player coordinating ER stress response-related gene expression. ATF3 superinduction was due to increased turnover of ATF3 transcript via stabilization with HuR protein in the cancer cells under ER stress. Moreover, enhanced ATF3 caused inhibitory action against ER stress-induced cancer chemokines that are potent mediators determining the survival and metastatic potential of epithelial cancer cells. Although enhanced ATF3 was a negative regulator of the well known proinflammatory transcription factor NF-κB, blocking of NF-κB signaling did not affect ER stress-induced chemokine expression. Instead, immediately expressed transcription factor early growth response protein 1 (EGR-1) was positively involved in cancer chemokine induction by ER stressors. ER stress-induced EGR-1 and subsequent chemokine production were repressed by ATF3. Mechanistically, ATF3 directly interacted with and recruited HDAC1 protein, which led to epigenetic suppression of EGR-1 expression and subsequent chemokine production. Conclusively, superinduced ATF3 attenuated ER stress-induced cancer chemokine expression by epigenetically interfering with induction of EGR-1, a transcriptional modulator crucial to cancer chemokine production. Thus, these results suggest a potent therapeutic intervention of ER stress response-related cancer-favoring events by ATF3.
Collapse
Affiliation(s)
- Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea,; Research Institute for Basic Sciences and Medical Research Institute, Pusan National University, Busan 609-735, Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Kee Hun Do
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Jiyeon Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Chang Gyu Oh
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Hye Jin Choi
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Bo Gyoung Song
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Seung Joon Lee
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | - Yong Sik Kim
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul 110-799, Korea, and
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 626-870, Korea,; Research Institute for Basic Sciences and Medical Research Institute, Pusan National University, Busan 609-735, Korea,; Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Busan 609-735, South Korea.
| |
Collapse
|
43
|
Kim J, Kwak HJ, Cha JY, Jeong YS, Rhee SD, Kim KR, Cheon HG. Metformin suppresses lipopolysaccharide (LPS)-induced inflammatory response in murine macrophages via activating transcription factor-3 (ATF-3) induction. J Biol Chem 2014; 289:23246-23255. [PMID: 24973221 DOI: 10.1074/jbc.m114.577908] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Metformin, a well known antidiabetic agent that improves peripheral insulin sensitivity, also elicits anti-inflammatory actions, but its mechanism is unclear. Here, we investigated the mechanism responsible for the anti-inflammatory effect of metformin action in lipopolysaccharide (LPS)-stimulated murine macrophages. Metformin inhibited LPS-induced production of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in a concentration-dependent manner and in parallel induction of activating transcription factor-3 (ATF-3), a transcription factor and member of the cAMP-responsive element-binding protein family. ATF-3 knockdown abolished the inhibitory effects of metformin on LPS-induced proinflammatory cytokine production accompanied with reversal of metformin-induced suppression of mitogen-activated protein kinase (MAPK) phosphorylation. Conversely, AMP-activated protein kinase (AMPK) phosphorylation and NF-κB suppression by metformin were unaffected by ATF-3 knockdown. ChIP-PCR analysis revealed that LPS-induced NF-κB enrichments on the promoters of IL-6 and TNF-α were replaced by ATF-3 upon metformin treatment. AMPK knockdown blunted all the effects of metformin (ATF-3 induction, proinflammatory cytokine inhibition, and MAPK inactivation), suggesting that AMPK activation by metformin is required for and precedes ATF-3 induction. Oral administration of metformin to either mice with LPS-induced endotoxemia or ob/ob mice lowered the plasma and tissue levels of TNF-α and IL-6 and increased ATF-3 expression in spleen and lungs. These results suggest that metformin exhibits anti-inflammatory action in macrophages at least in part via pathways involving AMPK activation and ATF-3 induction.
Collapse
Affiliation(s)
- Juyoung Kim
- Department of Pharmacology and Pharmaceutical Sciences and Gachon University, Incheon 406-799, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Pharmacology and Pharmaceutical Sciences and Gachon University, Incheon 406-799, Republic of Korea
| | - Ji-Young Cha
- Department of Molecular Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-799, Republic of Korea
| | - Yun-Seung Jeong
- Department of Molecular Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-799, Republic of Korea
| | - Sang Dahl Rhee
- Bioorganic Science Division, Korea Research Institute of Chemical Technology, Taejeon, 305-343, Republic of Korea, and
| | - Kwang Rok Kim
- Bioorganic Science Division, Korea Research Institute of Chemical Technology, Taejeon, 305-343, Republic of Korea, and
| | - Hyae Gyeong Cheon
- Department of Pharmacology and Pharmaceutical Sciences and Gachon University, Incheon 406-799, Republic of Korea; Gachon Medical Research Institute, Gil Medical Center, Incheon 405-760, Republic of Korea.
| |
Collapse
|
44
|
The regulatory role of activating transcription factor 2 in inflammation. Mediators Inflamm 2014; 2014:950472. [PMID: 25049453 PMCID: PMC4090481 DOI: 10.1155/2014/950472] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 05/30/2014] [Indexed: 01/06/2023] Open
Abstract
Activating transcription factor 2 (ATF2) is a member of the leucine zipper family of DNA-binding proteins and is widely distributed in tissues including the liver, lung, spleen, and kidney. Like c-Jun and c-Fos, ATF2 responds to stress-related stimuli and may thereby influence cell proliferation, inflammation, apoptosis, oncogenesis, neurological development and function, and skeletal remodeling. Recent studies clarify the regulatory role of ATF2 in inflammation and describe potential inhibitors of this protein. In this paper, we summarize the properties and functions of ATF2 and explore potential applications of ATF2 inhibitors as tools for research and for the development of immunosuppressive and anti-inflammatory drugs.
Collapse
|
45
|
Toedebusch RG, Roberts MD, Wells KD, Company JM, Kanosky KM, Padilla J, Jenkins NT, Perfield JW, Ibdah JA, Booth FW, Rector RS. Unique transcriptomic signature of omental adipose tissue in Ossabaw swine: a model of childhood obesity. Physiol Genomics 2014; 46:362-75. [PMID: 24642759 DOI: 10.1152/physiolgenomics.00172.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
To better understand the impact of childhood obesity on intra-abdominal adipose tissue phenotype, a complete transcriptomic analysis using deep RNA-sequencing (RNA-seq) was performed on omental adipose tissue (OMAT) obtained from lean and Western diet-induced obese juvenile Ossabaw swine. Obese animals had 88% greater body mass, 49% greater body fat content, and a 60% increase in OMAT adipocyte area (all P < 0.05) compared with lean pigs. RNA-seq revealed a 37% increase in the total transcript number in the OMAT of obese pigs. Ingenuity Pathway Analysis showed transcripts in obese OMAT were primarily enriched in the following categories: 1) development, 2) cellular function and maintenance, and 3) connective tissue development and function, while transcripts associated with RNA posttranslational modification, lipid metabolism, and small molecule biochemistry were reduced. DAVID and Gene Ontology analyses showed that many of the classically recognized gene pathways associated with adipose tissue dysfunction in obese adults including hypoxia, inflammation, angiogenesis were not altered in OMAT in our model. The current study indicates that obesity in juvenile Ossabaw swine is characterized by increases in overall OMAT transcript number and provides novel data describing early transcriptomic alterations that occur in response to excess caloric intake in visceral adipose tissue in a pig model of childhood obesity.
Collapse
Affiliation(s)
| | | | - Kevin D Wells
- Animal Sciences, University of Missouri, Columbia, Missouri
| | | | - Kayla M Kanosky
- Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Child Health, University of Missouri, Columbia, Missouri; Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | - James W Perfield
- Department of Food Science, University of Missouri, Columbia, Missouri; Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Jamal A Ibdah
- Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri; Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Frank W Booth
- Biomedical Sciences, University of Missouri, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - R Scott Rector
- Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri; Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; Research Service, Harry S. Truman Memorial VA Medical Center, University of Missouri, Columbia, Missouri; and
| |
Collapse
|
46
|
Iwasaki Y, Suganami T, Hachiya R, Shirakawa I, Kim-Saijo M, Tanaka M, Hamaguchi M, Takai-Igarashi T, Nakai M, Miyamoto Y, Ogawa Y. Activating transcription factor 4 links metabolic stress to interleukin-6 expression in macrophages. Diabetes 2014; 63:152-61. [PMID: 23990363 DOI: 10.2337/db13-0757] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic inflammation is a molecular element of the metabolic syndrome and type 2 diabetes. Saturated fatty acids (SFAs) are considered to be an important proinflammatory factor. However, it is still incompletely understood how SFAs induce proinflammatory cytokine expression. Hereby we report that activating transcription factor (ATF) 4, a transcription factor that is induced downstream of metabolic stresses including endoplasmic reticulum (ER) stress, plays critical roles in SFA-induced interleukin-6 (Il6) expression. DNA microarray analysis using primary macrophages revealed that the ATF4 pathway is activated by SFAs. Haploinsufficiency and short hairpin RNA-based knockdown of ATF4 in macrophages markedly inhibited SFA- and metabolic stress-induced Il6 expression. Conversely, pharmacological activation of the ATF4 pathway and overexpression of ATF4 resulted in enhanced Il6 expression. Moreover, ATF4 acts in synergy with the Toll-like receptor-4 signaling pathway, which is known to be activated by SFAs. At a molecular level, we found that ATF4 exerts its proinflammatory effects through at least two different mechanisms: ATF4 is involved in SFA-induced nuclear factor-κB activation; and ATF4 directly activates the Il6 promoter. These findings provide evidence suggesting that ATF4 links metabolic stress and Il6 expression in macrophages.
Collapse
Affiliation(s)
- Yorihiro Iwasaki
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
NF-κB1 inhibits NOD2-induced cytokine secretion through ATF3-dependent mechanisms. Mol Cell Biol 2013; 33:4857-71. [PMID: 24100018 DOI: 10.1128/mcb.00797-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Regulation of microbially induced cytokine secretion is critical in intestinal immune homeostasis. NOD2, the Crohn's disease-associated bacterial peptidoglycan sensor, activates the NF-κB pathway. After chronic NOD2 stimulation in human macrophages, cytokine secretion is significantly attenuated, similar to the situation in the intestinal environment. We find that NF-κB1 (p105/p50) expression is upregulated with chronic NOD2 stimulation and is required for attenuation of cytokine secretion in vitro in human macrophages and in vivo in mice. Upon chronic NOD2 stimulation, regulation of both activating (H3K4Me2 and H4Ac) and inhibitory (H3K27Me3) histone modifications was observed within cytokine gene promoters; these outcomes were NF-κB1 dependent. In addition to enhanced binding to cytokine gene promoters with chronic NOD2 stimulation, NF-κB1 bound to the promoter of the transcriptional repressor, ATF3. ATF3 was then induced and bound to cytokine gene promoters; both features were impaired upon NF-κB1 knockdown. Restoring ATF3 expression under NF-κB1 knockdown conditions restored NOD2-mediated cytokine downregulation. Finally, NF-κB1 and ATF3 cooperate with other inhibitory pathways, including IRAKM and secreted mediators, to downregulate cytokine secretion after chronic NOD2 stimulation. Therefore, we identify NF-κB1 and ATF3 as critical mechanisms through which NOD2 downregulates cytokines and contributes to intestinal immune homeostasis.
Collapse
|
48
|
Park SH, Do KH, Choi HJ, Kim J, Kim KH, Park J, Oh CG, Moon Y. Novel regulatory action of ribosomal inactivation on epithelial Nod2-linked proinflammatory signals in two convergent ATF3-associated pathways. THE JOURNAL OF IMMUNOLOGY 2013; 191:5170-81. [PMID: 24098051 DOI: 10.4049/jimmunol.1301145] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In response to excessive nucleotide-binding oligomerization domain-containing protein 2 (Nod2) stimulation caused by mucosal bacterial components, gut epithelia need to activate regulatory machinery to maintain epithelial homeostasis. Activating transcription factor 3 (ATF3) is a representative regulator in the negative feedback loop that modulates TLR-associated inflammatory responses. In the current study, the regulatory effects of ribosomal stress-induced ATF3 on Nod2-stimulated proinflammatory signals were assessed. Ribosomal inactivation caused persistent ATF3 expression that in turn suppressed proinflammatory chemokine production facilitated by Nod2. Decreased chemokine production was due to attenuation of Nod2-activated NF-κB and early growth response protein 1 (EGR-1) signals by ATF3. However, the underlying molecular mechanisms involve two convergent regulatory pathways. Although ATF3 induced by ribosomal inactivation regulated Nod2-induced EGR-1 expression epigenetically through the recruitment of histone deacetylase 1, NF-κB regulation was associated with posttranscriptional regulation by ATF3 rather than epigenetic modification. ATF3 induced by ribosomal inactivation led to the destabilization of p65 mRNA caused by nuclear entrapment of transcript-stabilizing human Ag R protein via direct interaction with ATF3. These findings demonstrate that ribosomal stress-induced ATF3 is a critical regulator in the convergent pathways between EGR-1 and NF-κB, which contributes to the suppression of Nod2-activated proinflammatory gene expression.
Collapse
Affiliation(s)
- Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Microbiology and Immunology, Medical Research Institute, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
A Taiwanese Propolis Derivative Induces Apoptosis through Inducing Endoplasmic Reticular Stress and Activating Transcription Factor-3 in Human Hepatoma Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:658370. [PMID: 24222778 PMCID: PMC3814109 DOI: 10.1155/2013/658370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 09/01/2013] [Indexed: 12/22/2022]
Abstract
Activating transcription factor-(ATF-) 3, a stress-inducible transcription factor, is rapidly upregulated under various stress conditions and plays an important role in inducing cancer cell apoptosis. NBM-TP-007-GS-002 (GS-002) is a Taiwanese propolin G (PPG) derivative. In this study, we examined the antitumor effects of GS-002 in human hepatoma Hep3B and HepG2 cells in vitro. First, we found that GS-002 significantly inhibited cell proliferation and induced cell apoptosis in dose-dependent manners. Several main apoptotic indicators were found in GS-002-treated cells, such as the cleaved forms of caspase-3, caspase-9, and poly(ADP-ribose) polymerase (PARP). GS-002 also induced endoplasmic reticular (ER) stress as evidenced by increases in ER stress-responsive proteins including glucose-regulated protein 78 (GRP78), growth arrest- and DNA damage-inducible gene 153 (GADD153), phosphorylated eukaryotic initiation factor 2α (eIF2α), phosphorylated protein endoplasmic-reticular-resident kinase (PERK), and ATF-3. The induction of ATF-3 expression was mediated by mitogen-activated protein kinase (MAPK) signaling pathways in GS-002-treated cells. Furthermore, we found that GS-002 induced more cell apoptosis in ATF-3-overexpressing cells. These results suggest that the induction of apoptosis by the propolis derivative, GS-002, is partially mediated through ER stress and ATF-3-dependent pathways, and GS-002 has the potential for development as an antitumor drug.
Collapse
|
50
|
Snodgrass RG, Huang S, Choi IW, Rutledge JC, Hwang DH. Inflammasome-mediated secretion of IL-1β in human monocytes through TLR2 activation; modulation by dietary fatty acids. THE JOURNAL OF IMMUNOLOGY 2013; 191:4337-47. [PMID: 24043885 DOI: 10.4049/jimmunol.1300298] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many studies have shown that TLR4- and TLR2-deficient mice are protected from high-fat diet-induced inflammation and insulin resistance, suggesting that saturated fatty acids derived from the high-fat diet activate TLR-mediated proinflammatory signaling pathways and induce insulin resistance. However, evidence that palmitic acid, the major dietary saturated fatty acid, can directly activate TLR has not been demonstrated. In this article, we present multiple lines of evidence showing that palmitic acid directly activates TLR2, a major TLR expressed on human monocytes, by inducing heterodimerization with TLR1 in an NADPH oxidase-dependent manner. Dimerization of TLR2 with TLR1 was inhibited by the n-3 fatty acid docosahexaenoic acid. Activation of TLR2 by palmitic acid leads to expression of pro-IL-1β that is cleaved by caspase-1, which is constitutively present in monocytes, to release mature IL-1β. Our results reveal mechanistic insight about how palmitic acid activates TLR2, upregulates NALP3 expression, and induces inflammasome-mediated IL-1β production in human monocytes, which can trigger enhanced inflammation in peripheral tissues, and suggest that these processes are dynamically modulated by the types of dietary fat we consume.
Collapse
Affiliation(s)
- Ryan G Snodgrass
- U.S. Department of Agriculture, Agricultural Research Service Western Human Nutrition Research Center, Davis, CA 95616
| | | | | | | | | |
Collapse
|