1
|
Stapornwongkul KS, Hahn E, Poliński P, Salamó Palau L, Arató K, Yao L, Williamson K, Gritti N, Anlas K, Osuna Lopez M, Patil KR, Heemskerk I, Ebisuya M, Trivedi V. Glycolytic activity instructs germ layer proportions through regulation of Nodal and Wnt signaling. Cell Stem Cell 2025; 32:744-758.e7. [PMID: 40245870 PMCID: PMC12048219 DOI: 10.1016/j.stem.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 10/29/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Abstract
Metabolic pathways can influence cell fate decisions, yet their regulative role during embryonic development remains poorly understood. Here, we demonstrate an instructive role of glycolytic activity in regulating signaling pathways involved in mesoderm and endoderm specification. Using a mouse embryonic stem cell (mESC)-based in vitro model for gastrulation, we found that glycolysis inhibition increases ectodermal cell fates at the expense of mesodermal and endodermal lineages. We demonstrate that this relationship is dose dependent, enabling metabolic control of germ layer proportions through exogenous glucose levels. We further show that glycolysis acts as an upstream regulator of Nodal and Wnt signaling and that its influence on cell fate specification can be decoupled from its effects on growth. Finally, we confirm the generality of our findings using a human gastrulation model. Our work underscores the dependence of signaling pathways on metabolic conditions and provides mechanistic insight into the nutritional regulation of cell fate decision-making.
Collapse
Affiliation(s)
- Kristina S Stapornwongkul
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain.
| | - Elisa Hahn
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Patryk Poliński
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Laura Salamó Palau
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Krisztina Arató
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - LiAng Yao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kate Williamson
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Nicola Gritti
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Kerim Anlas
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | | | - Kiran R Patil
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Idse Heemskerk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Miki Ebisuya
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain; Cluster of Excellence Physics of Life, TU Dresden, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| | - Vikas Trivedi
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain; Developmental Biology, EMBL Heidelberg, Heidelberg 69117, Germany.
| |
Collapse
|
2
|
Zhang W, Qi X, Han M, Jia Q, Li X, Yin W, Wang Y, Wu H, Shao H, Peng C, Su C, Sai L. Activation of Sirt1 by acetate alleviates silicofibrosis: Contribution of the gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117969. [PMID: 40020386 DOI: 10.1016/j.ecoenv.2025.117969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Silicosis is a prevalent occupational disease marked by progressive pulmonary fibrosis. Despite its significant health burden, the pathogenesis of silicosis remains unclear, and no specific therapeutic drugs are available. In this study, we developed a novel intervention strategy targeting gut microbiota and investigated its underlying mechanisms. Using 16S rRNA gene sequencing, we observed significant gut microbiota dysbiosis in silicosis rats at different times (1-8 weeks), notably characterized by altered relative abundance of Ruminococcus and Lactobacillus. Fecal microbiota transplantation altered the gut microbiota structure of silicosis rats, alleviated silica-induced lung histopathological injury, with LEfSe analysis identifying Bifidobacterium as a potential biomarker. Treatment with Bifidobacterium reduced the level of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and fibrosis markers (collagen III, α-SMA and vimentin) in the lungs of silicosis rats, accompanied with increased serum acetic acid levels. Acetate, a major metabolite of Bifidobacterium, demonstrated similar protective effects against silicosis in this study, suggesting its role as a key mediator of Bifidobacterium action in the lungs. Both Bifidobacterium and acetate significantly upregulated Sirt1 in intestinal and lung tissues, while Sirt1 inhibition diminished their benefits to silicosis. As a widely studied histone deacetylase, Sirt1 was proven to be markedly reduced in the lungs of silicosis rats in this study. EX-527, a potent Sirt1 inhibitor, could worsen silicosis damage by upregulating the level of TGF-β1 and the degree of Smad2/3 acetylation. Our study highlights the efficacy of postbiotics, such as Bifidobacterium and acetate, and identifies Sirt1 as a promising target for silicosis treatment.
Collapse
Affiliation(s)
- Weiliang Zhang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Xuejie Qi
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Mingming Han
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xixi Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenhui Yin
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanhui Wang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Heng Wu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Cheng Peng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China; Eusyn Institute of Health Science, Brisbane, QLD 4102, Australia; Shandong FMT Centre for Gut Microbiome Health, Jinan, Shandong, China
| | - Chongyi Su
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Linlin Sai
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
3
|
Daems M, Ponomarev LC, Simoes-Faria R, Nobis M, Scheele CLGJ, Luttun A, Ghesquière B, Zwijsen A, Jones EAV. Smad1/5 is acetylated in the dorsal aortae of the mouse embryo before the onset of blood flow, driving early arterial gene expression. Cardiovasc Res 2024; 120:2078-2091. [PMID: 39253943 DOI: 10.1093/cvr/cvae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 06/22/2024] [Indexed: 09/11/2024] Open
Abstract
AIMS During embryonic development, arteriovenous (AV) differentiation ensures proper blood vessel formation and maturation. Defects in arterial or venous identity cause inappropriate fusion of vessels, resulting in atypical shunts, so-called AV malformations (AVMs). Currently, the mechanism behind AVM formation remains unclear, and treatment options are fairly limited. Mammalian AV differentiation is initiated before the onset of blood flow in the embryo; however, this pre-flow mechanism is poorly understood. Here, we aimed to unravel the role of Smad1/5 signalling in pre-flow arterial identity and, in the process, uncovered an unexpected control mechanism of Smad1/5 signalling. METHODS AND RESULTS We establish that despite Notch1 being expressed in the pre-flow mouse embryo, it is not activated, nor is it necessary for the expression of the earliest arterial genes in the dorsal aortae (i.e. Hey1 and Gja4). Furthermore, interrupting blood flow by using the Ncx1 KO model completely prevents the activation of Notch1 signalling, suggesting a strong role of shear stress in maintaining arterial identity. We demonstrate that early expression of Hey1 and Gja4 requires SMAD1/5 signalling. Using embryo cultures, we show that Smad1/5 signalling is activated through the Alk1/Alk5/transforming growth factor (TGF)βR2 receptor complex, with TGFβ1 as a necessary ligand. Furthermore, our findings demonstrate that early arterial gene expression requires the acetylation of Smad1/5 proteins, rendering them more sensitive to TGFβ1 stimulation. Blocking acetyl-CoA production prevents pre-flow arterial expression of Hey1 and Gja4, while stabilizing acetylation rescues their expression. CONCLUSION Our findings highlight the importance of the acetyl-CoA production in the cell and provide a novel control mechanism of Smad1/5 signalling involving protein acetylation. As disturbed canonical Smad1/5 signalling is involved in several vascular conditions, our results offer new insights in treatment options for circumventing canonical Smad1/5 signalling.
Collapse
MESH Headings
- Animals
- Smad5 Protein/metabolism
- Smad5 Protein/genetics
- Smad1 Protein/metabolism
- Smad1 Protein/genetics
- Acetylation
- Gene Expression Regulation, Developmental
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Mice, Knockout
- Regional Blood Flow
- Signal Transduction
- Aorta/metabolism
- Aorta/physiopathology
- Aorta/embryology
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Protein Processing, Post-Translational
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Mice, Inbred C57BL
- Receptors, Transforming Growth Factor beta/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Mice
- Sodium-Calcium Exchanger
Collapse
Affiliation(s)
- Margo Daems
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Ljuba C Ponomarev
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Rita Simoes-Faria
- Metabolomics Expertise Centre, VIB Centre for Cancer Biology, 3000 Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, 3000 Leuven, Belgium
| | - Max Nobis
- Intravital Imaging Expertise Centre, VIB Centre for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Colinda L G J Scheele
- Department of Oncology, Laboratory for Intravital Imaging and Dynamics of Tumour Progression, VIB Centre for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Aernout Luttun
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Centre, VIB Centre for Cancer Biology, 3000 Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, 3000 Leuven, Belgium
| | - An Zwijsen
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
4
|
Runa F, Ortiz-Soto G, de Barros NR, Kelber JA. Targeting SMAD-Dependent Signaling: Considerations in Epithelial and Mesenchymal Solid Tumors. Pharmaceuticals (Basel) 2024; 17:326. [PMID: 38543112 PMCID: PMC10975212 DOI: 10.3390/ph17030326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024] Open
Abstract
SMADs are the canonical intracellular effector proteins of the TGF-β (transforming growth factor-β). SMADs translocate from plasma membrane receptors to the nucleus regulated by many SMAD-interacting proteins through phosphorylation and other post-translational modifications that govern their nucleocytoplasmic shuttling and subsequent transcriptional activity. The signaling pathway of TGF-β/SMAD exhibits both tumor-suppressing and tumor-promoting phenotypes in epithelial-derived solid tumors. Collectively, the pleiotropic nature of TGF-β/SMAD signaling presents significant challenges for the development of effective cancer therapies. Here, we review preclinical studies that evaluate the efficacy of inhibitors targeting major SMAD-regulating and/or -interacting proteins, particularly enzymes that may play important roles in epithelial or mesenchymal compartments within solid tumors.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| | | | | | - Jonathan A Kelber
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
- Department of Biology, Baylor University, Waco, TX 76706, USA
| |
Collapse
|
5
|
Jun JH, Kim JS, Palomera LF, Jo DG. Dysregulation of histone deacetylases in ocular diseases. Arch Pharm Res 2024; 47:20-39. [PMID: 38151648 DOI: 10.1007/s12272-023-01482-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
Ocular diseases are a growing global concern and have a significant impact on the quality of life. Cataracts, glaucoma, age-related macular degeneration, and diabetic retinopathy are the most prevalent ocular diseases. Their prevalence and the global market size are also increasing. However, the available pharmacotherapy is currently limited. These diseases share common pathophysiological features, including neovascularization, inflammation, and/or neurodegeneration. Histone deacetylases (HDACs) are a class of enzymes that catalyze the removal of acetyl groups from lysine residues of histone and nonhistone proteins. HDACs are crucial for regulating various cellular processes, such as gene expression, protein stability, localization, and function. They have also been studied in various research fields, including cancer, inflammatory diseases, neurological disorders, and vascular diseases. Our study aimed to investigate the relationship between HDACs and ocular diseases, to identify a new strategy for pharmacotherapy. This review article explores the role of HDACs in ocular diseases, specifically focusing on diabetic retinopathy, age-related macular degeneration, and retinopathy of prematurity, as well as optic nerve disorders, such as glaucoma and optic neuropathy. Additionally, we explore the interplay between HDACs and key regulators of fibrosis and angiogenesis, such as TGF-β and VEGF, highlighting the potential of targeting HDAC as novel therapeutic strategies for ocular diseases.
Collapse
Affiliation(s)
- Jae Hyun Jun
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
- Department of Pharmacology, CKD Research Institute, Chong Kun Dang Pharmaceutical Co., Yongin, 16995, Korea
| | - Jun-Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
| | - Leon F Palomera
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
6
|
Yang S, Yang G, Wang X, Xiang J, Kang L, Liang Z. SIRT2 alleviated renal fibrosis by deacetylating SMAD2 and SMAD3 in renal tubular epithelial cells. Cell Death Dis 2023; 14:646. [PMID: 37777567 PMCID: PMC10542381 DOI: 10.1038/s41419-023-06169-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/06/2023] [Accepted: 09/21/2023] [Indexed: 10/02/2023]
Abstract
Transforming growth factor-β (TGF-β) is the primary factor that drives fibrosis in most, if not all, forms of chronic kidney disease. In kidneys that are obstructed, specific deletion of Sirt2 in renal tubule epithelial cells (TEC) has been shown to aggravate renal fibrosis, while renal tubule specific overexpression of Sirt2 has been shown to ameliorate renal fibrosis. Similarly, specific deletion of Sirt2 in hepatocyte aggravated CCl4-induced hepatic fibrosis. In addition, we have demonstrated that SIRT2 overexpression and knockdown restrain and enhance TGF-β-induced fibrotic gene expression, respectively, in TEC. Mechanistically, SIRT2 reduced the phosphorylation, acetylation, and nuclear localization levels of SMAD2 and SMAD3, leading to inhibition of the TGF-β signaling pathway. Further studies have revealed that that SIRT2 was able to directly interact with and deacetylate SMAD2 at lysine 451, promoting its ubiquitination and degradation. Notably, loss of SMAD specific E3 ubiquitin protein ligase 2 abolishes the ubiquitination and degradation of SMAD2 induced by SIRT2 in SMAD2. Regarding SMAD3, we have found that SIRT2 interact with and deacetylates SMAD3 at lysine 341 and 378 only in the presence of TGF-β, thereby reducing its activation. This study provides initial indication of the anti-fibrotic role of SIRT2 in renal tubules and hepatocytes, suggesting its therapeutic potential for fibrosis.
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Guangyan Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Xinyu Wang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Jiaqing Xiang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Lin Kang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People's Hospital, Southern University of Science and Technology, Shenzhen, China.
| | - Zhen Liang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
7
|
Yi Y, Lan X, Li Y, Yan C, Lv J, Zhang T, Jiang W. Fatty acid synthesis and oxidation regulate human endoderm differentiation by mediating SMAD3 nuclear localization via acetylation. Dev Cell 2023; 58:1670-1687.e4. [PMID: 37516106 DOI: 10.1016/j.devcel.2023.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/02/2023] [Accepted: 07/07/2023] [Indexed: 07/31/2023]
Abstract
Metabolic remodeling is one of the earliest events that occur during cell differentiation. Here, we define fatty acid metabolism as a key player in definitive endoderm differentiation from human embryonic stem cells. Fatty acid β-oxidation is enhanced while lipogenesis is decreased, and this is due to the phosphorylation of lipogenic enzyme acetyl-CoA carboxylase by AMPK. More importantly, inhibition of fatty acid synthesis by either its inhibitors or AMPK agonist significantly promotes human endoderm differentiation, while blockade of fatty acid oxidation impairs differentiation. Mechanistically, reduced de novo fatty acid synthesis and enhanced fatty acid β-oxidation both contribute to the accumulation of intracellular acetyl-CoA, which guarantees the acetylation of SMAD3 and further causes nuclear localization to promote endoderm differentiation. Thus, our current study identifies a fatty acid synthesis/oxidation shift during early differentiation and presents an instructive role for fatty acid metabolism in regulating human endoderm differentiation.
Collapse
Affiliation(s)
- Ying Yi
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xianchun Lan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yinglei Li
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chenchao Yan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Jing Lv
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; College of Life Science, Cangzhou Normal University, Cangzhou 061000, China
| | - Tianzhe Zhang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
8
|
Luo W, Li Y, Zeng Y, Li Y, Cheng M, Zhang C, Li F, Wu Y, Huang C, Yang X, Kremerskothen J, Zhang J, Zhang C, Tu S, Li Z, Luo Z, Lin Z, Yan X. Tea domain transcription factor TEAD4 mitigates TGF-β signaling and hepatocellular carcinoma progression independently of YAP. J Mol Cell Biol 2023; 15:mjad010. [PMID: 36806855 PMCID: PMC10446140 DOI: 10.1093/jmcb/mjad010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 02/12/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023] Open
Abstract
Tea domain transcription factor 4 (TEAD4) plays a pivotal role in tissue development and homeostasis by interacting with Yes-associated protein (YAP) in response to Hippo signaling inactivation. TEAD4 and YAP can also cooperate with transforming growth factor-β (TGF-β)-activated Smad proteins to regulate gene transcription. Yet, it remains unclear whether TEAD4 plays a YAP-independent role in TGF-β signaling. Here, we unveil a novel tumor suppressive function of TEAD4 in liver cancer via mitigating TGF-β signaling. Ectopic TEAD4 inhibited TGF-β-induced signal transduction, Smad transcriptional activity, and target gene transcription, consequently suppressing hepatocellular carcinoma cell proliferation and migration in vitro and xenograft tumor growth in mice. Consistently, depletion of endogenous TEAD4 by siRNAs enhanced TGF-β signaling in cancer cells. Mechanistically, TEAD4 associates with receptor-regulated Smads (Smad2/3) and Smad4 in the nucleus, thereby impairing the binding of Smad2/3 to the histone acetyltransferase p300. Intriguingly, these negative effects of TEAD4 on TGF-β/Smad signaling are independent of YAP, as impairing the TEAD4-YAP interaction through point mutagenesis or depletion of YAP and/or its paralog TAZ has little effect. Together, these results unravel a novel function of TEAD4 in fine tuning TGF-β signaling and liver cancer progression in a YAP-independent manner.
Collapse
Affiliation(s)
- Weicheng Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Yi Li
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
| | - Yi Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Yining Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Minzhang Cheng
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Cheng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Fei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Yiqing Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Chunhong Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Joachim Kremerskothen
- Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster 48149, Germany
| | - Jianmin Zhang
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14203, USA
| | - Chunbo Zhang
- School of Pharmacy, Nanchang
University Jiangxi Medical College, Nanchang 330008, China
| | - Shuo Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
| | - Zhihua Li
- Key Laboratory of Breast Diseases of Jiangxi Province, Nanchang People’s Hospital, Nanchang 330025, China
| | - Zhijun Luo
- Department of Pathology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330006, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 405200, China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University Jiangxi Medical College, Nanchang 330031, China
- Key Laboratory of Breast Diseases of Jiangxi Province, Nanchang People’s Hospital, Nanchang 330025, China
| |
Collapse
|
9
|
Pharmacological inhibition of HDAC6 improves muscle phenotypes in dystrophin-deficient mice by downregulating TGF-β via Smad3 acetylation. Nat Commun 2022; 13:7108. [PMID: 36402791 PMCID: PMC9675748 DOI: 10.1038/s41467-022-34831-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy.
Collapse
|
10
|
The Polyunsaturated Fatty Acids, EPA and DHA, Ameliorate Myocardial Infarction-induced Heart Failure by Inhibiting p300-HAT Activity in Rats. J Nutr Biochem 2022; 106:109031. [DOI: 10.1016/j.jnutbio.2022.109031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/24/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022]
|
11
|
Lim Y, Jeong A, Kwon DH, Lee YU, Kim YK, Ahn Y, Kook T, Park WJ, Kook H. P300/CBP-Associated Factor Activates Cardiac Fibroblasts by SMAD2 Acetylation. Int J Mol Sci 2021; 22:9944. [PMID: 34576109 PMCID: PMC8472677 DOI: 10.3390/ijms22189944] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/31/2021] [Accepted: 09/10/2021] [Indexed: 01/07/2023] Open
Abstract
Various heart diseases cause cardiac remodeling, which in turn leads to ineffective contraction. Although it is an adaptive response to injury, cardiac fibrosis contributes to this remodeling, for which the reactivation of quiescent myofibroblasts is a key feature. In the present study, we investigated the role of the p300/CBP-associated factor (PCAF), a histone acetyltransferase, in the activation of cardiac fibroblasts. An intraperitoneal (i.p.) injection of a high dose (160 mg/kg) of isoproterenol (ISP) induced cardiac fibrosis and reduced the amount of the PCAF in cardiac fibroblasts in the mouse heart. However, the PCAF activity was significantly increased in cardiac fibroblasts, but not in cardiomyocytes, obtained from ISP-administered mice. An in vitro study using human cardiac fibroblast cells recapitulated the in vivo results; an treatment with transforming growth factor-β1 (TGF-β1) reduced the PCAF, whereas it activated the PCAF in the fibroblasts. PCAF siRNA attenuated the TGF-β1-induced increase in and translocation of fibrosis marker proteins. PCAF siRNA blocked TGF-β1-mediated gel contraction and cell migration. The PCAF directly interacted with and acetylated mothers against decapentaplegic homolog 2 (SMAD2). PCAF siRNA prevented TGF-β1-induced phosphorylation and the nuclear localization of SMAD2. These results suggest that the increase in PCAF activity during cardiac fibrosis may participate in SMAD2 acetylation and thereby in its activation.
Collapse
Affiliation(s)
- Yongwoon Lim
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.L.); (A.J.); (D.-H.K.); (Y.-U.L.); (T.K.)
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.-K.K.); (Y.A.)
- BK21 Plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju 61469, Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| | - Anna Jeong
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.L.); (A.J.); (D.-H.K.); (Y.-U.L.); (T.K.)
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.-K.K.); (Y.A.)
- BK21 Plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju 61469, Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| | - Duk-Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.L.); (A.J.); (D.-H.K.); (Y.-U.L.); (T.K.)
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.-K.K.); (Y.A.)
- BK21 Plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju 61469, Korea
| | - Yeong-Un Lee
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.L.); (A.J.); (D.-H.K.); (Y.-U.L.); (T.K.)
- Health and Environment Research Institute of Gwangju, 584, Mujin-daero, Seo-gu, Gwangju 61954, Korea
| | - Young-Kook Kim
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.-K.K.); (Y.A.)
- BK21 Plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju 61469, Korea
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea
| | - Youngkeun Ahn
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.-K.K.); (Y.A.)
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61469, Korea
| | - Taewon Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.L.); (A.J.); (D.-H.K.); (Y.-U.L.); (T.K.)
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Woo-Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.L.); (A.J.); (D.-H.K.); (Y.-U.L.); (T.K.)
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Korea; (Y.-K.K.); (Y.A.)
- BK21 Plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju 61469, Korea
| |
Collapse
|
12
|
Kim SM, Hur WH, Kang BY, Lee SW, Roh PR, Park DJ, Sung PS, Yoon SK. Death-Associated Protein 6 (Daxx) Alleviates Liver Fibrosis by Modulating Smad2 Acetylation. Cells 2021; 10:1742. [PMID: 34359912 PMCID: PMC8305094 DOI: 10.3390/cells10071742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/27/2022] Open
Abstract
Transforming growth factor-β (TGF-β) has been identified as an inducer of hepatocyte epithelial-mesenchymal transition (EMT), which triggers liver fibrosis. Death-associated protein 6 (Daxx) is known to be associated with the TGF-β-induced apoptotic pathway, but the function of Daxx in liver fibrosis remains unknown. This study aimed to elucidate the role of Daxx in liver fibrosis. We used liver fibrosis tissues from humans and mice to assess Daxx expression. EMT properties and TGF-β signaling pathway activation were investigated in the Daxx-overexpressing FL83B cell line. The therapeutic effect of Daxx was investigated in a mouse model of liver fibrosis by the hydrodynamic injection of plasmids. The expression of Daxx was markedly decreased in hepatocytes from fibrotic human and mouse livers, as well as in hepatocytes treated with TGF-β in vitro. The overexpression of Daxx inhibited the EMT process by interfering with the TGF-β-induced phosphorylation of Smad2. Coimmunoprecipitation analysis confirmed that Daxx reduced the transcriptional activity of Smad2 by binding to its MH1 domain and interfering with Smad2 acetylation. In addition, the therapeutic delivery of Daxx alleviated liver fibrosis in a thioacetamide-induced fibrosis mouse model. Overall, our results indicate that Daxx could be a potential therapeutic target to modulate fibrogenesis, as well as a useful biomarker for liver fibrosis.
Collapse
Affiliation(s)
- Sung-Min Kim
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
| | - Won-Hee Hur
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
| | - Byung-Yoon Kang
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
| | - Sung-Won Lee
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - Pu-Reun Roh
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
| | - Dong-Jun Park
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
| | - Pil-Soo Sung
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - Seung-Kew Yoon
- The Catholic University Liver Research Centre, Department of Biomedicine & Health Sciences, POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.-M.K.); (W.-H.H.); (B.-Y.K.); (S.-W.L.); (P.-R.R.); (D.-J.P.); (P.-S.S.)
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Korea
| |
Collapse
|
13
|
FOXO3a Protects against Kidney Injury in Type II Diabetic Nephropathy by Promoting Sirt6 Expression and Inhibiting Smad3 Acetylation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5565761. [PMID: 34122724 PMCID: PMC8172321 DOI: 10.1155/2021/5565761] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/11/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN) is the most common cause of end-stage renal disease. Although numerous reports have demonstrated a correlation between epithelial-mesenchymal transition (EMT) and renal fibrosis, how these processes lead to tubular dysfunction remains unclear. Here, we show that FOXO3a protects kidneys from injury in type II DN by increasing Sirt6 expression, which deacetylates Smad3 and inhibits its transcriptional activity. The results showed that progressive EMT in the kidneys from db/db mice is associated with Sirt6 downregulation and involved in tubular injury and dysfunction. The reduction of Sirt6 levels in db/db mice resulted in progressive kidney injury, indicating the protective role of Sirt6. Furthermore, Sirt6 was shown to directly bind to Smad3, a key downstream mediator of TGF-β, and could deacetylate it to inhibit its nuclear accumulation and transcriptional activity in HK2 cells. Besides, we demonstrate that FOXO3a activates Sirt6 expression by binding to its promoter. shRNA-induced FOXO3a knockdown in the kidneys of db/db mice exacerbated tubular injury and renal function loss. Mechanistically, FOXO3a protects against kidney injury in type II DN through the Sirt6/Smad3 axis. Thus, the pharmacological targeting of FOXO3a-mediated Sirt6/Smad3 signaling pathways may provide a novel strategy for treating type II DN.
Collapse
|
14
|
Zhang J, Li Y, Liu Q, Huang Y, Li R, Wu T, Zhang Z, Zhou J, Huang H, Tang Q, Huang C, Zhao Y, Zhang G, Jiang W, Mo L, Zhang J, Xie W, He J. Sirt6 Alleviated Liver Fibrosis by Deacetylating Conserved Lysine 54 on Smad2 in Hepatic Stellate Cells. Hepatology 2021; 73:1140-1157. [PMID: 32535965 PMCID: PMC8048913 DOI: 10.1002/hep.31418] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/06/2020] [Accepted: 05/20/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUNDS AND AIMS Activation of hepatic stellate cells (HSCs) is a central driver of fibrosis. This study aimed to elucidate the role of the deacetylase sirtuin 6 (Sirt6) in HSC activation and liver fibrosis. APPROACH AND RESULTS Gain-of-function and loss-of-function models were used to study the function of Sirt6 in HSC activation. Mass spectrometry was used to determine the specific acetylation site. The lecithin retinol acyltransferase-driven cyclization recombination recombinase construct (CreERT2) mouse line was created to generate HSC-specific conditional Sirt6-knockout mice (Sirt6△HSC ). We found that Sirt6 is most abundantly expressed in HSCs as compared with other liver cell types. The expression of Sirt6 was decreased in activated HSCs and fibrotic livers of mice and humans. Sirt6 knockdown and Sirt6 overexpression increased and decreased fibrogenic gene expression, respectively, in HSCs. Mechanistically, Sirt6 inhibited the phosphorylation and nuclear localization of mothers against decapentaplegic homolog (Smad) 2. Further study demonstrated that Sirt6 could directly interact with Smad2, deacetylate Smad2, and decrease the transcription of transforming growth factor β/Smad2 signaling. Mass spectrometry revealed that Sirt6 deacetylated conserved lysine 54 on Smad2. Mutation of lysine 54 to Arginine in Smad2 abolished the regulatory effect of Sirt6. In vivo, specific ablation of Sirt6 in HSCs exacerbated hepatocyte injury and cholestasis-induced liver fibrosis in mice. With targeted delivery of the Sirt6 agonist MDL-800, its concentration was 9.28-fold higher in HSCs as compared with other liver cells and alleviated hepatic fibrosis. CONCLUSIONS Sirt6 plays a key role in HSC activation and liver fibrosis by deacetylating the profibrogenic transcription factor Smad2. Sirt6 may be a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Jinhang Zhang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Ya Huang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Rui Li
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Tong Wu
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Zijing Zhang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Jian Zhou
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Hui Huang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Qin Tang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Cuiyuan Huang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Yingnan Zhao
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Guorong Zhang
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| | - Wei Jiang
- Molecular Medicine Research CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Li Mo
- Center of Gerontology and Geriatrics WestChina Hospital of Sichuan UniversityChengduChina
| | - Jian Zhang
- Department of PathophysiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao-Tong University School of Medicine (SJTU-SM)ShanghaiChina
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical SciencesUniversity of PittsburghPittsburghPA
| | - Jinhan He
- Department of PharmacyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina.,Laboratory of Clinical Pharmacy and Adverse Drug ReactionWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
15
|
Sinha A, Iyengar PV, ten Dijke P. E3 Ubiquitin Ligases: Key Regulators of TGFβ Signaling in Cancer Progression. Int J Mol Sci 2021; 22:E476. [PMID: 33418880 PMCID: PMC7825147 DOI: 10.3390/ijms22020476] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor β (TGFβ) is a secreted growth and differentiation factor that influences vital cellular processes like proliferation, adhesion, motility, and apoptosis. Regulation of the TGFβ signaling pathway is of key importance to maintain tissue homeostasis. Perturbation of this signaling pathway has been implicated in a plethora of diseases, including cancer. The effect of TGFβ is dependent on cellular context, and TGFβ can perform both anti- and pro-oncogenic roles. TGFβ acts by binding to specific cell surface TGFβ type I and type II transmembrane receptors that are endowed with serine/threonine kinase activity. Upon ligand-induced receptor phosphorylation, SMAD proteins and other intracellular effectors become activated and mediate biological responses. The levels, localization, and function of TGFβ signaling mediators, regulators, and effectors are highly dynamic and regulated by a myriad of post-translational modifications. One such crucial modification is ubiquitination. The ubiquitin modification is also a mechanism by which crosstalk with other signaling pathways is achieved. Crucial effector components of the ubiquitination cascade include the very diverse family of E3 ubiquitin ligases. This review summarizes the diverse roles of E3 ligases that act on TGFβ receptor and intracellular signaling components. E3 ligases regulate TGFβ signaling both positively and negatively by regulating degradation of receptors and various signaling intermediates. We also highlight the function of E3 ligases in connection with TGFβ's dual role during tumorigenesis. We conclude with a perspective on the emerging possibility of defining E3 ligases as drug targets and how they may be used to selectively target TGFβ-induced pro-oncogenic responses.
Collapse
Affiliation(s)
| | | | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (A.S.); (P.V.I.)
| |
Collapse
|
16
|
Wu M, Xu H, Liu J, Tan X, Wan S, Guo M, Long Y, Xu Y. Metformin and Fibrosis: A Review of Existing Evidence and Mechanisms. J Diabetes Res 2021; 2021:6673525. [PMID: 34007848 PMCID: PMC8102119 DOI: 10.1155/2021/6673525] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/16/2022] Open
Abstract
Fibrosis is a physiological response to organ injury and is characterized by the excessive deposition of connective tissue components in an organ, which results in the disruption of physiological architecture and organ remodeling, ultimately leading to organ failure and death. Fibrosis in the lung, kidney, and liver accounts for a substantial proportion of the global burden of disability and mortality. To date, there are no effective therapeutic strategies for controlling fibrosis. A class of metabolically targeted chemicals, such as adenosine monophosphate-activated protein kinase (AMPK) activators and peroxisome proliferator-activated receptor (PPAR) agonists, shows strong potential in fighting fibrosis. Metformin, which is a potent AMPK activator and is the only recommended first-line drug for the treatment of type 2 diabetes, has emerged as a promising method of fibrosis reduction or reversion. In this review, we first summarize the key experimental and clinical studies that have specifically investigated the effects of metformin on organ fibrosis. Then, we discuss the mechanisms involved in mediating the antifibrotic effects of metformin in depth.
Collapse
Affiliation(s)
- Maoyan Wu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| | - Huiwen Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| | - Jingyu Liu
- Southwest Medical University, Luzhou, Sichuan, China 646000
| | - Xiaozhen Tan
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| | - Shengrong Wan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| | - Man Guo
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China 646000
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China 646000
| |
Collapse
|
17
|
Aspirin inhibits TGFβ2-induced epithelial to mesenchymal transition of lens epithelial cells: selective acetylation of K56 and K122 in histone H3. Biochem J 2020; 477:75-97. [PMID: 31815277 DOI: 10.1042/bcj20190540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/04/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022]
Abstract
Posterior capsule opacification (PCO) is a complication after cataract surgery that can disrupt vision. The epithelial to mesenchymal transition (EMT) of lens epithelial cells (LECs) in response to transforming growth factor β2 (TGFβ2) has been considered an obligatory mechanism for PCO. In this study, we tested the efficacy of aspirin in inhibiting the TGFβ2-mediated EMT of human LECs, LECs in human lens capsular bags, and lensectomized mice. In human LECs, the levels of the EMT markers α-smooth muscle actin (α-SMA) and fibronectin were drastically reduced by treatment with 2 mM aspirin. Aspirin also halted the EMT response of TGFβ2 when introduced after EMT initiation. In human capsular bags, treatment with 2 mM aspirin significantly suppressed posterior capsule wrinkling and the expression α-SMA in capsule-adherent LECs. The inhibition of TGFβ2-mediated EMT in human LECs was not dependent on Smad phosphorylation or MAPK and AKT-mediated signaling. We found that aspirin significantly increased the acetylation of K56 and K122 in histone H3 of human LECs. Chromatin immunoprecipitation assays using acetyl-H3K56 or acetyl-H3K122 antibody revealed that aspirin blocked the TGFβ2-induced acetylation of H3K56 and H3K122 at the promoter regions of ACTA2 and COL1A1. After lensectomy in mice, we observed an increase in the proliferation and α-SMA expression of the capsule-adherent LECs, which was ameliorated by aspirin administration through drinking water. Taken together, our results showed that aspirin inhibits TGFβ2-mediated EMT of LECs, possibly from epigenetic down-regulation of EMT-related genes.
Collapse
|
18
|
Concise Review: The Regulatory Mechanism of Lysine Acetylation in Mesenchymal Stem Cell Differentiation. Stem Cells Int 2020; 2020:7618506. [PMID: 32399051 PMCID: PMC7204305 DOI: 10.1155/2020/7618506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/02/2020] [Indexed: 12/30/2022] Open
Abstract
Nowadays, the use of MSCs has attracted considerable attention in the global science and technology field, with the self-renewal and multidirectional differentiation potential for diabetes, obesity treatment, bone repair, nerve repair, myocardial repair, and so on. Epigenetics plays an important role in the regulation of mesenchymal stem cell differentiation, which has become a research hotspot in the medical field. This review focuses on the role of lysine acetylation modification on the determination of MSC differentiation direction. During this progress, the recruitment of lysine acetyltransferases (KATs) and lysine deacetylases (KDACs) is the crux of transcriptional mechanisms in the dynamic regulation of key genes controlling MSC multidirectional differentiation.
Collapse
|
19
|
TGFβ2-induced formation of lipid droplets supports acidosis-driven EMT and the metastatic spreading of cancer cells. Nat Commun 2020; 11:454. [PMID: 31974393 PMCID: PMC6978517 DOI: 10.1038/s41467-019-14262-3] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 12/23/2019] [Indexed: 11/10/2022] Open
Abstract
Acidosis, a common characteristic of the tumor microenvironment, is associated with alterations in metabolic preferences of cancer cells and progression of the disease. Here we identify the TGF-β2 isoform at the interface between these observations. We document that acidic pH promotes autocrine TGF-β2 signaling, which in turn favors the formation of lipid droplets (LD) that represent energy stores readily available to support anoikis resistance and cancer cell invasiveness. We find that, in cancer cells of various origins, acidosis-induced TGF-β2 activation promotes both partial epithelial-to-mesenchymal transition (EMT) and fatty acid metabolism, the latter supporting Smad2 acetylation. We show that upon TGF-β2 stimulation, PKC-zeta-mediated translocation of CD36 facilitates the uptake of fatty acids that are either stored as triglycerides in LD through DGAT1 or oxidized to generate ATP to fulfill immediate cellular needs. We also address how, by preventing fatty acid mobilization from LD, distant metastatic spreading may be inhibited. The tumour microenvironment is known to have an acidic pH but how this influences cancer cell phenotype is unclear. Here, the authors show that tumour cells upregulate TGF-β2 under acidosis, which leads to the increased formation of lipid droplets allowing for invasiveness and metastases.
Collapse
|
20
|
Wang J, Wu M, Zheng D, Zhang H, Lv Y, Zhang L, Tan HS, Zhou H, Lao YZ, Xu HX. Garcinol inhibits esophageal cancer metastasis by suppressing the p300 and TGF-β1 signaling pathways. Acta Pharmacol Sin 2020; 41:82-92. [PMID: 31371781 PMCID: PMC7471459 DOI: 10.1038/s41401-019-0271-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023]
Abstract
Metastasis causes the main lethality in esophageal cancer patient. Garcinol, a natural compound extracted from Gambogic genera, is a histone acetyltransferase (HAT) inhibitor that has shown anticancer activities such as cell cycle arrest and apoptosis induction. In this study, we investigated the effects of garcinol on the metastasis of esophageal cancer in vitro and in vivo. We found that garcinol (5-15 μM) dose-dependently inhibited the migration and invasion of human esophageal cancer cell lines KYSE150 and KYSE450 in wound healing, transwell migration, and Matrigel invasion assays. Furthermore, garcinol treatment dose-dependently decreased the protein levels of p300/CBP (transcriptional cofactors and HATs) and p-Smad2/3 expression in the nucleus, thus impeding tumor cell proliferation and metastasis. Knockdown of p300 could inhibit cell metastasis, but CBP knockdown did not affect the cell mobility. It has been reported that TGF-β1 stimulated the phosphorylation of Smad2/3, which directly interact with p300/CBP in the nucleus, and upregulating HAT activity of p300. We showed that garcinol treatment dose-dependently suppressed TGF-β1-activated Smad and non-Smad pathway, inhibiting esophageal cancer cell metastasis. In a tail vein injection pulmonary metastasis mouse model, intraperitoneal administration of garcinol (20 mg/kg) or 5-FU (20 mg/kg) significantly decreased the number of lung tumor nodules and the expression levels of Ki-67, p300, and p-Smad2/3 in lung tissues. In conclusion, our study demonstrates that garcinol inhibits esophageal cancer metastasis in vitro and in vivo, which might be related to the suppression of p300 and TGF-β1 signaling pathways, suggesting the therapeutic potential of Garcinol for metastatic tumors.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- E1A-Associated p300 Protein/deficiency
- E1A-Associated p300 Protein/metabolism
- Esophageal Neoplasms/drug therapy
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/pathology
- Garcinia/chemistry
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Signal Transduction/drug effects
- Terpenes/chemistry
- Terpenes/isolation & purification
- Terpenes/pharmacology
- Transforming Growth Factor beta1/metabolism
- Tumor Cells, Cultured
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Jing Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Man Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Lv
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong-Sheng Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan-Zhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hong-Xi Xu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
21
|
Cell-Based Mechanosensation, Epigenetics, and Non-Coding RNAs in Progression of Cardiac Fibrosis. Int J Mol Sci 2019; 21:ijms21010028. [PMID: 31861579 PMCID: PMC6982012 DOI: 10.3390/ijms21010028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/11/2019] [Accepted: 12/15/2019] [Indexed: 12/22/2022] Open
Abstract
The heart is par excellence the 'in-motion' organ in the human body. Compelling evidence shows that, besides generating forces to ensure continuous blood supply (e.g., myocardial contractility) or withstanding passive forces generated by flow (e.g., shear stress on endocardium, myocardial wall strain, and compression strain at the level of cardiac valves), cells resident in the heart respond to mechanical cues with the activation of mechanically dependent molecular pathways. Cardiac stromal cells, most commonly named cardiac fibroblasts, are central in the pathologic evolution of the cardiovascular system. In their normal function, these cells translate mechanical cues into signals that are necessary to renew the tissues, e.g., by continuously rebuilding the extracellular matrix being subjected to mechanical stress. In the presence of tissue insults (e.g., ischemia), inflammatory cues, or modifiable/unmodifiable risk conditions, these mechanical signals may be 'misinterpreted' by cardiac fibroblasts, giving rise to pathology programming. In fact, these cells are subject to changing their phenotype from that of matrix renewing to that of matrix scarring cells-the so-called myo-fibroblasts-involved in cardiac fibrosis. The links between alterations in the abilities of cardiac fibroblasts to 'sense' mechanical cues and molecular pathology programming are still under investigation. On the other hand, various evidence suggests that cell mechanics may control stromal cells phenotype by modifying the epigenetic landscape, and this involves specific non-coding RNAs. In the present contribution, we will provide examples in support of this more integrated vision of cardiac fibrotic progression based on the decryption of mechanical cues in the context of epigenetic and non-coding RNA biology.
Collapse
|
22
|
Dewidar B, Meyer C, Dooley S, Meindl-Beinker N. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019; 8:cells8111419. [PMID: 31718044 PMCID: PMC6912224 DOI: 10.3390/cells8111419] [Citation(s) in RCA: 520] [Impact Index Per Article: 86.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is an advanced liver disease condition, which could progress to cirrhosis and hepatocellular carcinoma. To date, there is no direct approved antifibrotic therapy, and current treatment is mainly the removal of the causative factor. Transforming growth factor (TGF)-β is a master profibrogenic cytokine and a promising target to treat fibrosis. However, TGF-β has broad biological functions and its inhibition induces non-desirable side effects, which override therapeutic benefits. Therefore, understanding the pleiotropic effects of TGF-β and its upstream and downstream regulatory mechanisms will help to design better TGF-β based therapeutics. Here, we summarize recent discoveries and milestones on the TGF-β signaling pathway related to liver fibrosis and hepatic stellate cell (HSC) activation, emphasizing research of the last five years. This comprises impact of TGF-β on liver fibrogenesis related biological processes, such as senescence, metabolism, reactive oxygen species generation, epigenetics, circadian rhythm, epithelial mesenchymal transition, and endothelial-mesenchymal transition. We also describe the influence of the microenvironment on the response of HSC to TGF-β. Finally, we discuss new approaches to target the TGF-β pathway, name current clinical trials, and explain promises and drawbacks that deserve to be adequately addressed.
Collapse
Affiliation(s)
- Bedair Dewidar
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, 31527 Tanta, Egypt
| | - Christoph Meyer
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
| | - Nadja Meindl-Beinker
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
- Correspondence: ; Tel.: +49-621-383-4983; Fax: +49-621-383-1467
| |
Collapse
|
23
|
Higgins CE, Tang J, Mian BM, Higgins SP, Gifford CC, Conti DJ, Meldrum KK, Samarakoon R, Higgins PJ. TGF-β1-p53 cooperativity regulates a profibrotic genomic program in the kidney: molecular mechanisms and clinical implications. FASEB J 2019; 33:10596-10606. [PMID: 31284746 PMCID: PMC6766640 DOI: 10.1096/fj.201900943r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease affects >15% of the U.S. population and >850 million individuals worldwide. Fibrosis is the common outcome of many chronic renal disorders and, although the etiology varies (i.e., diabetes, hypertension, ischemia, acute injury, and urologic obstructive disorders), persistently elevated renal TGF-β1 levels result in the relentless progression of fibrotic disease. TGF-β1 orchestrates the multifaceted program of renal fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery and redifferentiation, and subsequent tubulointerstitial fibrosis, eventually leading to chronic renal disease. Recent findings implicate p53 as a cofactor in the TGF-β1-induced signaling pathway and a transcriptional coregulator of several TGF-β1 profibrotic response genes by complexing with receptor-activated SMADs, which are homologous to the small worms (SMA) and Drosophilia mothers against decapentaplegic (MAD) gene families. The cooperative p53-TGF-β1 genomic cluster includes genes involved in cell growth control and extracellular matrix remodeling [e.g., plasminogen activator inhibitor-1 (PAI-1; serine protease inhibitor, clade E, member 1), connective tissue growth factor, and collagen I]. Although the molecular basis for this codependency is unclear, many TGF-β1-responsive genes possess p53 binding motifs. p53 up-regulation and increased p53 phosphorylation; moreover, they are evident in nephrotoxin- and ischemia/reperfusion-induced injury, diabetic nephropathy, ureteral obstructive disease, and kidney allograft rejection. Pharmacologic and genetic approaches that target p53 attenuate expression of the involved genes and mitigate the fibrotic response, confirming a key role for p53 in renal disorders. This review focuses on mechanisms whereby p53 functions as a transcriptional regulator within the TGF-β1 cluster with an emphasis on the potent fibrosis-promoting PAI-1 gene.-Higgins, C. E., Tang, J., Mian, B. M., Higgins, S. P., Gifford, C. C., Conti, D. J., Meldrum, K. K., Samarakoon, R., Higgins, P. J. TGF-β1-p53 cooperativity regulates a profibrotic genomic program in the kidney: molecular mechanisms and clinical implications.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, New York, USA
- Division of Urology, Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Kirstan K. Meldrum
- Division of Pediatric Urology, Central Michigan University, Mount Pleasant, Michigan, USA
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
- The Urological Institute of Northeastern New York, Albany, New York, USA
- Division of Urology, Department of Surgery, Albany Medical College, Albany, New York, USA
| |
Collapse
|
24
|
Jiang R, Zhou Y, Wang S, Pang N, Huang Y, Ye M, Wan T, Qiu Y, Pei L, Jiang X, Huang Y, Yang H, Ling W, Li X, Zhang Z, Yang L. Nicotinamide riboside protects against liver fibrosis induced by CCl 4 via regulating the acetylation of Smads signaling pathway. Life Sci 2019; 225:20-28. [PMID: 30928408 DOI: 10.1016/j.lfs.2019.03.064] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/25/2019] [Accepted: 03/25/2019] [Indexed: 12/28/2022]
Abstract
AIMS Increasing nicotinamide adenine dinucleotide (NAD+) by Nicotinamide riboside (NR) provides protective benefits in multiple disorders. However, the role of NR on liver fibrosis is unclear. We performed in vivo and in vitro experiments to test the hepatic protective effects of NR against liver fibrosis and the underlying mechanisms. MATERIALS AND METHODS Mice were injected with CCl4 to establish liver fibrosis model. NR was given by gavage to explore the hepatic protection of NR. LX-2 cells were given a TGF-β stimulation ± NR, the activation of LX-2 cells and the acetylation of Smads were analyzed. To further confirm the role of Sirt1 on the protective pathway of NR, we knockdown Sirt1 in LX-2 cells. KEY FINDINGS We found NR could prevent liver fibrosis and reverse the existing liver fibrosis. NR inhibited the activation of LX-2 cells induced by TGF-β, activated Sirt1 and deacetylated Smad2/3. Sirt1 knockdown diminished the inhibiting effect of NR on LX-2 cells activation, and increased expressions of acetylated Smads. In conclusion, NR could prevent liver fibrosis via suppressing activation of hepatic stellate cells (HSCs). This protective effect was mediated by regulating the acetylation of Smads signaling pathway. SIGNIFICANCE NR protected mice against liver fibrosis induced by CCl4. NR suppressed activation of hepatic stellate cells induced by TGF-β. NR protects liver fibrosis via increasing the activity of Sirt1 and decreasing the expression of P300, resulting in the deacetylation of Smads in stellate cells.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China; Nutrition Clinic, The Maternal & Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530003, People's Republic of China
| | - Yujia Zhou
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Sufan Wang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Nengzhi Pang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Yuanling Huang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Mingtong Ye
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Ting Wan
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Yun Qiu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Lei Pei
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Xuye Jiang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Yufeng Huang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510260, People's Republic of China
| | - Hainan Yang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510260, People's Republic of China
| | - Wenhua Ling
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China
| | - Xufeng Li
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510260, People's Republic of China.
| | - Zhenfeng Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510260, People's Republic of China.
| | - Lili Yang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, People's Republic of China.
| |
Collapse
|
25
|
Systemic Activation of Activin A Signaling Causes Chronic Kidney Disease-Mineral Bone Disorder. Int J Mol Sci 2018; 19:ijms19092490. [PMID: 30142896 PMCID: PMC6163495 DOI: 10.3390/ijms19092490] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 11/19/2022] Open
Abstract
The high cardiovascular mortality associated with chronic kidney disease (CKD) is caused in part by the CKD-mineral bone disorder (CKD-MBD) syndrome. The CKD-MBD consists of skeletal, vascular and cardiac pathology caused by metabolic derangements produced by kidney disease. The prevalence of osteopenia/osteoporosis resulting from the skeletal component of the CKD-MBD, renal osteodystrophy (ROD), in patients with CKD exceeds that of the general population and is a major public health concern. That CKD is associated with compromised bone health is widely accepted, yet the mechanisms underlying impaired bone metabolism in CKD are not fully understood. Therefore, clarification of the molecular mechanisms by which CKD produces ROD is of crucial significance. We have shown that activin A, a member of the transforming growth factor (TGF)-β super family, is an important positive regulator of receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis with Smad-mediated signaling being crucial for inducing osteoclast development and function. Recently, we have demonstrated systemic activation of activin receptors and activin A levels in CKD mouse models, such as diabetic CKD and Alport (AL) syndrome. In these CKD mouse models, bone remodeling caused by increased osteoclast numbers and activated osteoclastic bone resorption was observed and treatment with an activin receptor ligand trap repaired CKD-induced-osteoclastic bone resorption and stimulated individual osteoblastic bone formation, irrespective of parathyroid hormone (PTH) elevation. These findings have opened a new field for exploring mechanisms of activin A-enhanced osteoclast formation and function in CKD. Activin A appears to be a strong candidate for CKD-induced high-turnover ROD. Therefore, the treatment with the decoy receptor for activin A might be a good candidate for treatment for CKD-induced osteopenia or osteoporosis, indicating that the new findings from in these studies will lead to the identification of novel therapeutic targets for CKD-related and osteopenia and osteoporosis in general. In this review, we describe the impact of CKD-induced Smad signaling in osteoclasts, osteoblasts and vascular cells in CKD.
Collapse
|
26
|
Rios Garcia M, Steinbauer B, Srivastava K, Singhal M, Mattijssen F, Maida A, Christian S, Hess-Stumpp H, Augustin HG, Müller-Decker K, Nawroth PP, Herzig S, Berriel Diaz M. Acetyl-CoA Carboxylase 1-Dependent Protein Acetylation Controls Breast Cancer Metastasis and Recurrence. Cell Metab 2017; 26:842-855.e5. [PMID: 29056512 DOI: 10.1016/j.cmet.2017.09.018] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/18/2017] [Accepted: 09/20/2017] [Indexed: 02/08/2023]
Abstract
Breast tumor recurrence and metastasis represent the main causes of cancer-related death in women, and treatments are still lacking. Here, we define the lipogenic enzyme acetyl-CoA carboxylase (ACC) 1 as a key player in breast cancer metastasis. ACC1 phosphorylation was increased in invading cells both in murine and human breast cancer, serving as a point of convergence for leptin and transforming growth factor (TGF) β signaling. ACC1 phosphorylation was mediated by TGFβ-activated kinase (TAK) 1, and ACC1 inhibition was indispensable for the elevation of cellular acetyl-CoA, the subsequent increase in Smad2 transcription factor acetylation and activation, and ultimately epithelial-mesenchymal transition and metastasis induction. ACC1 deficiency worsened tumor recurrence upon primary tumor resection in mice, and ACC1 phosphorylation levels correlated with metastatic potential in breast and lung cancer patients. Given the demonstrated effectiveness of anti-leptin receptor antibody treatment in halting ACC1-dependent tumor invasiveness, our work defines a "metabolocentric" approach in metastatic breast cancer therapy.
Collapse
Affiliation(s)
- Marcos Rios Garcia
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; Technical University Munich, 85764 Neuherberg, Germany; Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany
| | - Brigitte Steinbauer
- Core Facility Tumor Models, German Cancer Research Center (DKFZ) and Medical Faculty Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Kshitij Srivastava
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ) and Medical Faculty Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ) and Medical Faculty Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Frits Mattijssen
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; Technical University Munich, 85764 Neuherberg, Germany; Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany
| | - Adriano Maida
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; Technical University Munich, 85764 Neuherberg, Germany; Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany
| | - Sven Christian
- Division Tumor Metabolism and Hypoxia, Bayer Health Care, 13353 Berlin, Germany
| | - Holger Hess-Stumpp
- Division Tumor Metabolism and Hypoxia, Bayer Health Care, 13353 Berlin, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ) and Medical Faculty Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Karin Müller-Decker
- Core Facility Tumor Models, German Cancer Research Center (DKFZ) and Medical Faculty Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Peter P Nawroth
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; Technical University Munich, 85764 Neuherberg, Germany; Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; Technical University Munich, 85764 Neuherberg, Germany; Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany.
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; Technical University Munich, 85764 Neuherberg, Germany; Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany.
| |
Collapse
|
27
|
García-Vizcaíno EM, Liarte S, Alonso-Romero JL, Nicolás FJ. Sirt1 interaction with active Smad2 modulates transforming growth factor-β regulated transcription. Cell Commun Signal 2017; 15:50. [PMID: 29187201 PMCID: PMC5706420 DOI: 10.1186/s12964-017-0205-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Background The simplicity of Transforming Growth Factor ß (TGFβ) signaling pathway, linear and non-amplified, hardly sustains its variety of responses. This is often justified by the complex regulation showed by Smad proteins, TGFβ signaling intracellular transducers, object of post-translational modifications that modulate TGFβ-dependent transcription. Protein acetylation is emerging as a compelling mechanism affecting the activities of significant transcription factors, including p53, FOXO or NF-kB. Smad proteins might be controlled by this mechanism, implying that accessory factors capable of altering Smads-transcriptional complexes acetylation status and hence regulate TGFβ responses remain to be identified. Understanding this interaction may help in the assessment of TGFβ signaling outcomes, extending from healthy physiology to pathological conditions and cancer. Methods A two-hybrid chimera interacting system allowed to identify Sirt1, a NAD+ dependent type III histone deacetylase, as a novel Smad2 interactor. Several well stablished cellular models were applied to characterize this interaction by means of co-immunoprecipitation of tagged proteins and immuno-fluorescence staining. The occurrence of the interaction at Smad2 driven transcriptomic complexes was studied by means of DNA-pull-down and chromatin immunoprecipitation (ChIP), while its effects were assessed by protein over-expression and siRNA applied into a TGFβ-dependent reporter gene assay. Results The interaction was confirmed and observed to be enhanced upon Smad2 acetylation, a known feature of active and nuclear Smad2. However, Sirt1 did not play a major role in Smad2 deacetylation. Anti-Sirt1 ChIP showed increased recovery of promoter regions corresponding to Smad2-driven genes after TGFβ-stimulation, while its occurrence at Smad2-dependent transcriptomic complexes on DNA was found to effectively modulate gene expression. Conclusions Sirt1 presence on Smad2-driven TGFβ-dependent regulatory elements was detected and found to increase after TGFβ treatment. Moreover, Sirt1 overexpression resulted in a decrease of the activity of a Smad2-driven TGFβ-dependent reporter gene, while Sirt1 interference increased its activity. This would confirm the relevance of the discovered Sirt1-Smad2 interaction for the regulation of TGFβ-dependent gene transcription. Electronic supplementary material The online version of this article (10.1186/s12964-017-0205-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eva María García-Vizcaíno
- Laboratorio de Oncología Molecular y TGFβ, Instituto Murciano de Investigaciones Biosanitarias Arrixaca, El Palmar, Murcia, Spain
| | - Sergio Liarte
- Laboratorio de Oncología Molecular y TGFβ, Instituto Murciano de Investigaciones Biosanitarias Arrixaca, El Palmar, Murcia, Spain
| | - José Luis Alonso-Romero
- Servicio de Oncología, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | - Francisco José Nicolás
- Laboratorio de Oncología Molecular y TGFβ, Instituto Murciano de Investigaciones Biosanitarias Arrixaca, El Palmar, Murcia, Spain.
| |
Collapse
|
28
|
Higgins SP, Tang Y, Higgins CE, Mian B, Zhang W, Czekay RP, Samarakoon R, Conti DJ, Higgins PJ. TGF-β1/p53 signaling in renal fibrogenesis. Cell Signal 2017; 43:1-10. [PMID: 29191563 DOI: 10.1016/j.cellsig.2017.11.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/26/2017] [Accepted: 11/27/2017] [Indexed: 01/04/2023]
Abstract
Fibrotic disorders of the renal, pulmonary, cardiac, and hepatic systems are associated with significant morbidity and mortality. Effective therapies to prevent or curtail the advancement to organ failure, however, remain a major clinical challenge. Chronic kidney disease, in particular, constitutes an increasing medical burden affecting >15% of the US population. Regardless of etiology (diabetes, hypertension, ischemia, acute injury, urologic obstruction), persistently elevated TGF-β1 levels are causatively linked to the activation of profibrotic signaling networks and disease progression. TGF-β1 is the principal driver of renal fibrogenesis, a dynamic pathophysiologic process that involves tubular cell injury/apoptosis, infiltration of inflammatory cells, interstitial fibroblast activation and excess extracellular matrix synthesis/deposition leading to impaired kidney function and, eventually, to chronic and end-stage disease. TGF-β1 activates the ALK5 type I receptor (which phosphorylates SMAD2/3) as well as non-canonical (e.g., src kinase, EGFR, JAK/STAT, p53) pathways that collectively drive the fibrotic genomic program. Such multiplexed signal integration has pathophysiological consequences. Indeed, TGF-β1 stimulates the activation and assembly of p53-SMAD3 complexes required for transcription of the renal fibrotic genes plasminogen activator inhibitor-1, connective tissue growth factor and TGF-β1. Tubular-specific ablation of p53 in mice or pifithrin-α-mediated inactivation of p53 prevents epithelial G2/M arrest, reduces the secretion of fibrotic effectors and attenuates the transition from acute to chronic renal injury, further supporting the involvement of p53 in disease progression. This review focuses on the pathophysiology of TGF-β1-initiated renal fibrogenesis and the role of p53 as a regulator of profibrotic gene expression.
Collapse
Affiliation(s)
- Stephen P Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States.
| | - Yi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States.
| | - Craig E Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States.
| | - Badar Mian
- Department of Surgery, Albany Medical College, Albany, NY 12208, United States; The Urological Institute of Northeastern New York, Albany Medical College, Albany, NY 12208, United States.
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States.
| | - Ralf-Peter Czekay
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States.
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States.
| | - David J Conti
- Department of Surgery, Albany Medical College, Albany, NY 12208, United States; Division of Transplantation Surgery, Albany Medical College, Albany, NY 12208, United States.
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States; Department of Surgery, Albany Medical College, Albany, NY 12208, United States; The Urological Institute of Northeastern New York, Albany Medical College, Albany, NY 12208, United States.
| |
Collapse
|
29
|
Xu P, Lin X, Feng XH. Posttranslational Regulation of Smads. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a022087. [PMID: 27908935 DOI: 10.1101/cshperspect.a022087] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Transforming growth factor β (TGF-β) family signaling dictates highly complex programs of gene expression responses, which are extensively regulated at multiple levels and vary depending on the physiological context. The formation, activation, and destruction of two major functional complexes in the TGF-β signaling pathway (i.e., the TGF-β receptor complexes and the Smad complexes that act as central mediators of TGF-β signaling) are direct targets for posttranslational regulation. Dysfunction of these complexes often leads or contributes to pathogenesis in cancer and fibrosis and in cardiovascular, and autoimmune diseases. Here we discuss recent insights into the roles of posttranslational modifications in the functions of the receptor-activated Smads in the common Smad4 and inhibitory Smads, and in the control of the physiological responses to TGF-β. It is now evident that these modifications act as decisive factors in defining the intensity and versatility of TGF-β responsiveness. Thus, the characterization of posttranslational modifications of Smads not only sheds light on how TGF-β controls physiological and pathological processes but may also guide us to manipulate the TGF-β responses for therapeutic benefits.
Collapse
Affiliation(s)
- Pinglong Xu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xia Lin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030
| | - Xin-Hua Feng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China.,Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
30
|
Epigenetics in fibrosis. Mol Aspects Med 2016; 54:89-102. [PMID: 27720780 DOI: 10.1016/j.mam.2016.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022]
Abstract
Fibrosis is a common and important disease. It is a pathological state due to excessive scar formation mediated by an increase in activated fibroblasts that express alpha smooth muscle actin and copious amounts of extracellular matrix molecules. Epigenetics is an area of research that encompasses three main mechanisms: methylation, histone modifications to the tails of histones and also non-coding RNAs including long and short non-coding RNAs. These three mechanisms all seek to regulate gene expression without a change in the underlying DNA sequence. In recent years an explosion of research, aided by deep sequencing technology becoming available, has demonstrated a role for epigenetics in fibrosis, either organ specific like lung fibrosis or more widespread as in systemic sclerosis. While the great majority of epigenetic work in fibrosis is centered on histone codes, more recently the non-coding RNAs have been examined in greater detail. It is known that one modification can affect the other and cross-talk among all three adds a new layer of complexity. This review aims to examine the role of epigenetics in fibrosis, evaluating all three mechanisms, and to suggest possible areas where epigenetics could be targeted therapeutically.
Collapse
|
31
|
Wang N, Xu Q, Tan HY, Hong M, Li S, Yuen MF, Feng Y. Berberine Inhibition of Fibrogenesis in a Rat Model of Liver Fibrosis and in Hepatic Stellate Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:8762345. [PMID: 27239214 PMCID: PMC4867075 DOI: 10.1155/2016/8762345] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/15/2016] [Indexed: 02/06/2023]
Abstract
Aim. To examine the effect of berberine (BBR) on liver fibrosis and its possible mechanisms through direct effects on hepatic stellate cells (HSC). Methods. The antifibrotic effect of BBR was determined in a rat model of bile duct ligation- (BDL-) induced liver fibrosis. Multiple cellular and molecular approaches were introduced to examine the effects of BBR on HSC. Results. BBR potently inhibited hepatic fibrosis induced by BDL in rats. It exhibited cytotoxicity to activated HSC at doses nontoxic to hepatocytes. High doses of BBR induced apoptosis of activated HSC, which was mediated by loss of mitochondrial membrane potential and Bcl-2/Bax imbalance. Low doses of BBR suppressed activation of HSC as evidenced by the inhibition of α-smooth muscle actin (α-SMA) expression and cell motility. BBR did not affect Smad2/3 phosphorylation but significantly activated 5' AMP-activated protein kinase (AMPK) signalling, which was responsible for the transcriptional inhibition by BBR of profibrogenic factors α-SMA and collagen in HSC. Conclusion. BBR is a promising agent for treating liver fibrosis through multiple mechanisms, at least partially by directly targeting HSC and by inhibiting the AMPK pathway. Its value as an antifibrotic drug in patients with liver disease deserves further investigation.
Collapse
Affiliation(s)
- Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Qihe Xu
- Centre for Integrative Chinese Medicine and Department of Renal Medicine, Faculty of Life Sciences and Medicine, King's College London, London SE5 9NU, UK
| | - Hor Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ming Hong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Man-Fung Yuen
- Division of Gastroenterology and Hepatology, Queen Mary Hospital and Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
32
|
Reid CD, Steiner AB, Yaklichkin S, Lu Q, Wang S, Hennessy M, Kessler DS. FoxH1 mediates a Grg4 and Smad2 dependent transcriptional switch in Nodal signaling during Xenopus mesoderm development. Dev Biol 2016; 414:34-44. [PMID: 27085753 DOI: 10.1016/j.ydbio.2016.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/28/2016] [Accepted: 04/06/2016] [Indexed: 02/07/2023]
Abstract
In the vertebrate blastula and gastrula the Nodal pathway is essential for formation of the primary germ layers and the organizer. Nodal autoregulatory feedback potentiates signaling activity, but mechanisms limiting embryonic Nodal ligand transcription are poorly understood. Here we describe a transcriptional switch mechanism mediated by FoxH1, the principle effector of Nodal autoregulation. FoxH1 contains a conserved engrailed homology (EH1) motif that mediates direct binding of groucho-related gene 4 (Grg4), a Groucho family corepressor. Nodal-dependent gene expression is suppressed by FoxH1, but enhanced by a FoxH1 EH1 mutant, indicating that the EH1 motif is necessary for repression. Grg4 blocks Nodal-induced mesodermal gene expression and Nodal autoregulation, suggesting that Grg4 limits Nodal pathway activity. Conversely, blocking Grg4 function in the ectoderm results in ectopic expression of Nodal target genes. FoxH1 and Grg4 occupy the Xnr1 enhancer, and Grg4 occupancy is dependent on the FoxH1 EH1 motif. Grg4 occupancy at the Xnr1 enhancer significantly decreases with Nodal activation or Smad2 overexpression, while FoxH1 occupancy is unaffected. These results suggest that Nodal-activated Smad2 physically displaces Grg4 from FoxH1, an essential feature of the transcriptional switch mechanism. In support of this model, when FoxH1 is unable to bind Smad2, Grg4 occupancy is maintained at the Xnr1 enhancer, even in the presence of Nodal signaling. Our findings reveal that FoxH1 mediates both activation and repression of Nodal gene expression. We propose that this transcriptional switch is essential to delimit Nodal pathway activity in vertebrate germ layer formation.
Collapse
Affiliation(s)
- Christine D Reid
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Aaron B Steiner
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Sergey Yaklichkin
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Qun Lu
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Shouwen Wang
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Morgan Hennessy
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Daniel S Kessler
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
SIRT1: A Novel Target for the Treatment of Muscular Dystrophies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6714686. [PMID: 27073590 PMCID: PMC4814699 DOI: 10.1155/2016/6714686] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/28/2016] [Indexed: 12/13/2022]
Abstract
Muscular dystrophies are inherited myogenic disorders accompanied by progressive skeletal muscle weakness and degeneration. Duchenne muscular dystrophy (DMD) is the most common and severe form of muscular dystrophy and is caused by mutations in the gene that encodes the cytoskeletal protein dystrophin. The treatment for DMD is limited to glucocorticoids, which are associated with multiple side effects. Thus, the identification of novel therapeutic targets is urgently needed. SIRT1 is an NAD+-dependent histone/protein deacetylase that plays roles in diverse cellular processes, including stress resistance and cell survival. Studies have shown that SIRT1 activation provides beneficial effects in the dystrophin-deficient mdx mouse, a model of DMD. SIRT1 activation leads to the attenuation of oxidative stress and inflammation, a shift from the fast to slow myofiber phenotype, and the suppression of tissue fibrosis. Although further research is needed to clarify the molecular mechanisms underlying the protective role of SIRT1 in mdx mice, we propose SIRT1 as a novel therapeutic target for patients with muscular dystrophies.
Collapse
|
34
|
Kaypee S, Sudarshan D, Shanmugam MK, Mukherjee D, Sethi G, Kundu TK. Aberrant lysine acetylation in tumorigenesis: Implications in the development of therapeutics. Pharmacol Ther 2016; 162:98-119. [PMID: 26808162 DOI: 10.1016/j.pharmthera.2016.01.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The 'language' of covalent histone modifications translates environmental and cellular cues into gene expression. This vast array of post-translational modifications on histones are more than just covalent moieties added onto a protein, as they also form a platform on which crucial cellular signals are relayed. The reversible lysine acetylation has emerged as an important post-translational modification of both histone and non-histone proteins, dictating numerous epigenetic programs within a cell. Thus, understanding the complex biology of lysine acetylation and its regulators is essential for the development of epigenetic therapeutics. In this review, we will attempt to address the complexities of lysine acetylation in the context of tumorigenesis, their role in cancer progression and emphasize on the modalities developed to target lysine acetyltransferases towards cancer treatment.
Collapse
Affiliation(s)
- Stephanie Kaypee
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
| | - Deepthi Sudarshan
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Debanjan Mukherjee
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India.
| |
Collapse
|
35
|
Aortic Remodelling Is Improved by 2,3,5,4'-Tetrahydroxystilbene-2-O-β-D-glucoside Involving the Smad3 Pathway in Spontaneously Hypertensive Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:789027. [PMID: 26693246 PMCID: PMC4677031 DOI: 10.1155/2015/789027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/15/2015] [Indexed: 12/04/2022]
Abstract
Hypertension is a common health problem that substantially increases the risk of cardiovascular disease. The condition increases blood pressure, which causes alterations in vascular structure and leads to the development of vascular pathologies. 2,3,5,4′-Tetrahydroxystilbene-2-O-β-D-glucoside (THSG), a resveratrol analogue extracted from a Chinese medicinal plant, has been proven to have numerous vascular protection functions. This study investigated whether THSG can improve vascular remodeling, which has thus far remained unclear. Orally administering THSG to spontaneously hypertensive rats (SHRs) aged 12 weeks for 14 weeks significantly inhibited intima-media thickness in the lower parts of the aortic arch, increased the vascular diastolic rate in response to acetylcholine, and reduced remodelling-related mRNA expression, such as that of ACTA2, CCL3, COL1A2, COL3A1, TIMP1 WISP2, IGFBP1, ECE1, KLF5, MYL1 BMP4, FN1, and PAI-1. Immunofluorescence staining also showed an inhibitory effect similar to that of THSG on PAI-1 protein expression in rat aortas. Results from immunoprecipitation and a Western blot assay showed that THSG inhibited the acetylation of Smad3. A chromatin immunoprecipitation assay showed that THSG prevented Smad3 binding to the PAI-1 proximal promoter in SHR aortas. In conclusion, our results demonstrated that the inhibitory effect of THSG on aortic remodelling involved the deacetylating role of Smad3 with increasing blood flow and with constant blood pressure.
Collapse
|
36
|
Su BH, Tseng YL, Shieh GS, Chen YC, Wu P, Shiau AL, Wu CL. Over-expression of prothymosin-α antagonizes TGFβ signalling to promote the development of emphysema. J Pathol 2015; 238:412-22. [DOI: 10.1002/path.4664] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 09/11/2015] [Accepted: 10/08/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Bing-Hua Su
- Department of Biochemistry and Molecular Biology, College of Medicine; National Cheng Kung University; Tainan Taiwan
| | - Yau-Lin Tseng
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine; National Cheng Kung University; Tainan Taiwan
| | - Gia-Shing Shieh
- Department of Urology; Tainan Hospital, Ministry of Health and Welfare; Tainan Taiwan
| | - Yi-Cheng Chen
- Department of Biochemistry and Molecular Biology, College of Medicine; National Cheng Kung University; Tainan Taiwan
| | - Pensee Wu
- Institute for Science and Technology in Medicine; Keele University; UK
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine; National Cheng Kung University; Tainan Taiwan
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine; National Cheng Kung University; Tainan Taiwan
| |
Collapse
|
37
|
von Bernhardi R, Cornejo F, Parada GE, Eugenín J. Role of TGFβ signaling in the pathogenesis of Alzheimer's disease. Front Cell Neurosci 2015; 9:426. [PMID: 26578886 PMCID: PMC4623426 DOI: 10.3389/fncel.2015.00426] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/09/2015] [Indexed: 12/19/2022] Open
Abstract
Aging is the main risk factor for Alzheimer’s disease (AD); being associated with conspicuous changes on microglia activation. Aged microglia exhibit an increased expression of cytokines, exacerbated reactivity to various stimuli, oxidative stress, and reduced phagocytosis of β-amyloid (Aβ). Whereas normal inflammation is protective, it becomes dysregulated in the presence of a persistent stimulus, or in the context of an inflammatory environment, as observed in aging. Thus, neuroinflammation can be a self-perpetuating deleterious response, becoming a source of additional injury to host cells in neurodegenerative diseases. In aged individuals, although transforming growth factor β (TGFβ) is upregulated, its canonical Smad3 signaling is greatly reduced and neuroinflammation persists. This age-related Smad3 impairment reduces protective activation while facilitating cytotoxic activation of microglia through several cellular mechanisms, potentiating microglia-mediated neurodegeneration. Here, we critically discuss the role of TGFβ-Smad signaling on the cytotoxic activation of microglia and its relevance in the pathogenesis of AD. Other protective functions, such as phagocytosis, although observed in aged animals, are not further induced by inflammatory stimuli and TGFβ1. Analysis in silico revealed that increased expression of receptor scavenger receptor (SR)-A, involved in Aβ uptake and cell activation, by microglia exposed to TGFβ, through a Smad3-dependent mechanism could be mediated by transcriptional co-factors Smad2/3 over the MSR1 gene. We discuss that changes of TGFβ-mediated regulation could at least partially mediate age-associated microglia changes, and, together with other changes on inflammatory response, could result in the reduction of protective activation and the potentiation of cytotoxicity of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Francisca Cornejo
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Guillermo E Parada
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Jaime Eugenín
- Laboratory of Neural Systems, Faculty of Chemistry and Biology, Department of Biology, Universidad de Santiago de Chile Santiago, Chile
| |
Collapse
|
38
|
Zhang Y, Wang S, Liu S, Li C, Wang J. Role of Smad signaling in kidney disease. Int Urol Nephrol 2015; 47:1965-75. [DOI: 10.1007/s11255-015-1115-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/18/2015] [Indexed: 01/21/2023]
|
39
|
Zhao LJ, Loewenstein PM, Green M. The adenovirus E1A oncoprotein N-terminal transcriptional repression domain enhances p300 autoacetylation and inhibits histone H3 Lys18 acetylation. Genes Cancer 2015; 6:30-7. [PMID: 25821559 PMCID: PMC4362482 DOI: 10.18632/genesandcancer.47] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/07/2015] [Indexed: 11/25/2022] Open
Abstract
Expression of the adenovirus E1A N-terminal transcription repression domain alone (E1A 1-80) represses transcription from specific promoters such as HER2 [1] and from reconstituted chromatin [2]. Significantly, E1A 1-80 can induce the death of human breast cancer cells over-expressing the HER2 oncogene [1] as well as other cancer cells. Here, we report that E1A 1-80 alone is sufficient to inhibit H3K18 acetylation in vivo and p300-mediated H3K18 acetylation in reconstituted chromatin. Of interest, hypoacetylation of H3K18 has been correlated with the survival of tumor cells and the poor prognosis of many cancers [3, 4]. E1A 1-80 enhances p300 autoacetylation and concurrently inhibits H3K18 acetylation in chromatin in a dose-dependent manner. Pre-acetylation of p300 by incubation with acetyl-CoA alone reduces p300's ability to acetylate H3K18 in chromatin. Additional acetylation of p300 in the presence of E1A 1-80 produces stronger inhibition of H3K18 acetylation. These findings indicate that autoacetylation of p300 greatly reduces its ability to acetylate H3K18. The results reported here combined with our previous findings suggest that E1A can repress transcription by multiple strategies, including altering the chromatin modifying activity of p300 and dissociating TFIID from the TATA box thus disrupting formation of the transcription pre-initiation complex [5, 6]
Collapse
Affiliation(s)
- Ling-Jun Zhao
- Institute for Molecular Virology, Saint Louis University School of Medicine, Doisy research Center, St. Louis, Missouri
| | - Paul M Loewenstein
- Institute for Molecular Virology, Saint Louis University School of Medicine, Doisy research Center, St. Louis, Missouri
| | - Maurice Green
- Institute for Molecular Virology, Saint Louis University School of Medicine, Doisy research Center, St. Louis, Missouri
| |
Collapse
|
40
|
Huang X, Xu J, Huang M, Li J, Dai L, Dai K, Zhang X. Histone deacetylase1 promotes TGF-β1-mediated early chondrogenesis through down-regulating canonical Wnt signaling. Biochem Biophys Res Commun 2014; 453:810-6. [DOI: 10.1016/j.bbrc.2014.10.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/07/2014] [Indexed: 12/22/2022]
|
41
|
Ahn J, Yoon Y, Yeu Y, Lee H, Park S. Impact of TGF-b on breast cancer from a quantitative proteomic analysis. Comput Biol Med 2013; 43:2096-102. [DOI: 10.1016/j.compbiomed.2013.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 09/22/2013] [Accepted: 09/25/2013] [Indexed: 02/04/2023]
|
42
|
Wang F, Marshall CB, Ikura M. Transcriptional/epigenetic regulator CBP/p300 in tumorigenesis: structural and functional versatility in target recognition. Cell Mol Life Sci 2013; 70:3989-4008. [PMID: 23307074 PMCID: PMC11113169 DOI: 10.1007/s00018-012-1254-4] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 11/08/2012] [Accepted: 12/20/2012] [Indexed: 01/19/2023]
Abstract
In eukaryotic cells, gene transcription is regulated by sequence-specific DNA-binding transcription factors that recognize promoter and enhancer elements near the transcriptional start site. Some coactivators promote transcription by connecting transcription factors to the basal transcriptional machinery. The highly conserved coactivators CREB-binding protein (CBP) and its paralog, E1A-binding protein (p300), each have four separate transactivation domains (TADs) that interact with the TADs of a number of DNA-binding transcription activators as well as general transcription factors (GTFs), thus mediating recruitment of basal transcription machinery to the promoter. Most promoters comprise multiple activator-binding sites, and many activators contain tandem TADs, thus multivalent interactions may stabilize CBP/p300 at the promoter, and intrinsically disordered regions in CBP/p300 and many activators may confer adaptability to these multivalent complexes. CBP/p300 contains a catalytic histone acetyltransferase (HAT) domain, which remodels chromatin to 'relax' its superstructure and enables transcription of proximal genes. The HAT activity of CBP/p300 also acetylates some transcription factors (e.g., p53), hence modulating the function of key transcriptional regulators. Through these numerous interactions, CBP/p300 has been implicated in complex physiological and pathological processes, and, in response to different signals, can drive cells towards proliferation or apoptosis. Dysregulation of the transcriptional and epigenetic functions of CBP/p300 is associated with leukemia and other types of cancer, thus it has been recognized as a potential anti-cancer drug target. In this review, we focus on recent exciting findings in the structural mechanisms of CBP/p300 involving multivalent and dynamic interactions with binding partners, which may pave new avenues for anti-cancer drug development.
Collapse
Affiliation(s)
- Feng Wang
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9 Canada
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7 Canada
- Present Address: Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Christopher B. Marshall
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9 Canada
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7 Canada
| | - Mitsuhiko Ikura
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9 Canada
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7 Canada
| |
Collapse
|
43
|
Gomez D, Kessler K, Michel JB, Vranckx R. Modifications of Chromatin Dynamics Control Smad2 Pathway Activation in Aneurysmal Smooth Muscle Cells. Circ Res 2013; 113:881-90. [DOI: 10.1161/circresaha.113.301989] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Rationale
:
The activation of the Smad2 signaling pathway is thought to play an important role in human aneurysmal diseases as described by an important body of research. We previously showed that constitutive Smad2 activation is associated with Smad2 mRNA overexpression in aneurysmal vascular smooth muscle cells (VSMCs), which is dependent on epigenetic regulation of the
SMAD2
promoter involving histone modifications. However, the underlying molecular mechanisms controlling Smad2 overexpression are currently unknown.
Objective
:
The aim of the present study is to understand the mechanisms regulating the constitutive Smad2 overexpression in VSMCs by identification of the histone-modifying enzymes, transcription factors, and cofactors responsible for Smad2 promoter activation in aneurysmal disease.
Methods and Results
:
This study was performed on medial tissue extracts and primary cultures of VSMCs of human thoracic aneurysms (n=17) and normal thoracic aortas (n=10). Here, we demonstrate that the activation of
SMAD2
promoter is driven by the recruitment of a multipartner complex, including the transcription factor p53 and histone acetyltransferases. Remarkably, the transcriptional regulatory network of the
SMAD2
promoter is dramatically altered in human aneurysmal VSMCs in vitro and in situ with a switch from Myc-dependent repression of
SMAD2
in normal vessel to a p53-dependent activation of
SMAD2
in aneurysms. Furthermore, histone acetyltransferases p300 and P300/CBP-associated protein play a major role in
SMAD2
promoter activation by acting on histone acetylation, p53 recruitment, and acetylation.
Conclusions
:
These results provide evidence for a major role of p53 and the complex composed of p300 and p300/CBP-associated protein in Smad2 activation in human aneurysmal VSMCs.
Collapse
Affiliation(s)
- Delphine Gomez
- From the INSERM, U698, Paris, France (D.G., K.K., J.-B.M., R.V.); and Université Paris Diderot, Sorbonne Paris Cité, Paris, France (D.G., K.K., J.-B.M., R.V.)
| | - Ketty Kessler
- From the INSERM, U698, Paris, France (D.G., K.K., J.-B.M., R.V.); and Université Paris Diderot, Sorbonne Paris Cité, Paris, France (D.G., K.K., J.-B.M., R.V.)
| | - Jean-Baptiste Michel
- From the INSERM, U698, Paris, France (D.G., K.K., J.-B.M., R.V.); and Université Paris Diderot, Sorbonne Paris Cité, Paris, France (D.G., K.K., J.-B.M., R.V.)
| | - Roger Vranckx
- From the INSERM, U698, Paris, France (D.G., K.K., J.-B.M., R.V.); and Université Paris Diderot, Sorbonne Paris Cité, Paris, France (D.G., K.K., J.-B.M., R.V.)
| |
Collapse
|
44
|
Activin stimulates CYP19A gene expression in human ovarian granulosa cell-like KGN cells via the Smad2 signaling pathway. Biochem Biophys Res Commun 2013; 436:443-8. [PMID: 23747729 DOI: 10.1016/j.bbrc.2013.05.124] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 05/29/2013] [Indexed: 11/21/2022]
Abstract
Activin, a transforming growth factor β family member, has a wide range of physiological roles during embryonic development and organogenesis. In the ovary, activin, secreted from ovarian granulosa cells, not only acts on the pituitary gland to regulate the gonadotropin secretion from the pituitary gland in an endocrine manner but also acts on granulosa cells in a paracrine/autocrine manner to regulate folliculogenesis. Previously, we showed that activin signals through activin type IB receptor (ActRIB) and up-regulates follicle-stimulating hormone receptor expression and P450 aromatase activity in human ovarian granulose cell-like KGN cells. In the current study, we demonstrate the direct involvement of Smad2 as a downstream signal mediator of ActRIB in the transcriptional regulation of the P450 aromatase gene (CYP19A) in KGN cells. Upon activin stimulation, Smad2 activation and an increase in P450 aromatase messenger RNA (mRNA) were observed in KGN cells. Interestingly, Smad2 phosphorylation correlated well with the increase in P450 aromatase mRNA. Reciprocally, knockdown of Smad2 mRNA in KGN cells led to a decrease in the P450 aromatase mRNA expression, suggesting that Smad2 regulates CYP19A gene expression. Further analysis of CYP19A promoter activity revealed that the 5' upstream region between -2069 and -1271bp is required for the activation by Smad2. Finally, we provide compelling evidence that Smad2 shows follicular stage-specific expression, which is high in granulosa cells of preantral or early antral follicles in mice. Our results suggest that activin signaling through the ActRIB-Smad2 pathway plays a pivotal role in CYP19A expression and thus in follicular development.
Collapse
|
45
|
Yuan H, Reddy MA, Sun G, Lanting L, Wang M, Kato M, Natarajan R. Involvement of p300/CBP and epigenetic histone acetylation in TGF-β1-mediated gene transcription in mesangial cells. Am J Physiol Renal Physiol 2013; 304:F601-13. [PMID: 23235480 PMCID: PMC3602713 DOI: 10.1152/ajprenal.00523.2012] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 12/08/2012] [Indexed: 01/01/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1)-induced expression of plasminogen activator inhibitor-1 (PAI-1) and p21 in renal mesangial cells (MCs) plays a major role in glomerulosclerosis and hypertrophy, key events in the pathogenesis of diabetic nephropathy. However, the involvement of histone acetyl transferases (HATs) and histone deacetylases (HDACs) that regulate epigenetic histone lysine acetylation, and their interaction with TGF-β1-responsive transcription factors, are not clear. We evaluated the roles of histone acetylation, specific HATs, and HDACs in TGF-β1-induced gene expression in rat mesangial cells (RMCs) and in glomeruli from diabetic mice. Overexpression of HATs CREB binding protein (CBP) or p300, but not p300/CBP-activating factor, significantly enhanced TGF-β1-induced PAI-1 and p21 mRNA levels as well as transactivation of their promoters in RMCs. Conversely, they were significantly attenuated by HAT domain mutants of CBP and p300 or overexpression of HDAC-1 and HDAC-5. Chromatin immunoprecipitation assays showed that TGF-β1 treatment led to a time-dependent enrichment of histone H3-lysine9/14-acetylation (H3K9/14Ac) and p300/CBP occupancies around Smad and Sp1 binding sites at the PAI-1 and p21 promoters. TGF-β1 also enhanced the interaction of p300 with Smad2/3 and Sp1 and increased Smad2/3 acetylation. High glucose-treated RMCs exhibited increased PAI-1 and p21 levels, and promoter H3K9/14Ac, which were blocked by TGF-β1 antibodies. Furthermore, increased PAI-1 and p21 expression was associated with elevated promoter H3K9/14Ac levels in glomeruli from diabetic mice. Thus TGF-β1-induced PAI-1 and p21 expression involves interaction of p300/CBP with Smads and Sp1, and increased promoter access via p300/CBP-induced H3K9/14Ac. This in turn can augment glomerular dysfunction linked to diabetic nephropathy.
Collapse
Affiliation(s)
- Hang Yuan
- Dept. of Diabetes, Beckman Research Institute of the City of Hope, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Ko H, So Y, Jeon H, Jeong MH, Choi HK, Ryu SH, Lee SW, Yoon HG, Choi KC. TGF-β1-induced epithelial-mesenchymal transition and acetylation of Smad2 and Smad3 are negatively regulated by EGCG in human A549 lung cancer cells. Cancer Lett 2013; 335:205-13. [PMID: 23419524 DOI: 10.1016/j.canlet.2013.02.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 01/22/2023]
Abstract
Transforming growth factor-β1, the key ligand of Smad-dependent signaling pathway, is critical for epithelial-mesenchymal transition during embryo-morphogenesis, fibrotic diseases, and tumor metastasis. In this study, we found that activation of p300/CBP, a histone acetyltransferase, by TGF-β1 mediates Epithelial-mesenchymal transition (EMT) via acetylating Smad2 and Smad3 in TGF-β1 signaling pathway. We demonstrated that treatment with EGCG inhibited p300/CBP activity in human lung cancer cells. Also, we observed that EGCG potently inhibited TGF-β1-induced EMT and reversed the up-regulation of various genes during EMT. Our findings suggest that EGCG inhibits the induction of p300/CBP activity by TGF-β1. Therefore, EGCG inhibits TGF-β1-mediated EMT by suppressing the acetylation of Smad2 and Smad3 in human lung cancer cells.
Collapse
Affiliation(s)
- Hyeonseok Ko
- Laboratory of Molecular Oncology, Cheil General Hospital & Women's Healthcare Center, Kwandong University College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ganglioside GM3 participates in the TGF-β1-induced epithelial-mesenchymal transition of human lens epithelial cells. Biochem J 2013; 449:241-51. [PMID: 23050851 DOI: 10.1042/bj20120189] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TGF-β (transforming growth factor-β)-induced EMT (epithelial-mesenchymal transition) induces the proliferation and migration of the HLE (human lens epithelial) cells. Ganglioside GM3, simple sialic-acid-containing glycosphingolipids on mammalian cell membranes, regulates various pathological phenomena such as insulin resistance and tumour progression. However, the relationship between ganglioside GM3 and TGF-β-induced EMT in the HLE B-3 cells is poorly understood. In the present study we demonstrated that ganglioside GM3 was involved in TGF-β1-induced EMT in HLE B-3 cells. Our results indicated that the expression of ganglioside GM3 and GM3 synthase mRNA were significantly increased in TGF-β1-induced HLE B-3 cells. Reporter gene analysis also demonstrated that transcriptional activation of the GM3 synthase gene was regulated by Sp1 (specificity protein 1) in HLE B-3 cells upon TGF-β1 stimulation. Interestingly, the inhibition of ganglioside GM3 expression by d-PDMP [d-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol] and GM3 synthase shRNA (short hairpin RNA) resulted significantly in the suppression of cell migration and EMT-related signalling in HLE B-3 cells stimulated by TGF-β. Furthermore, exogenous treatment of ganglioside GM3 rescued the expression of EMT molecules and cell migration suppressed by the depletion of ganglioside GM3 in TGF-β1-induced HLE B-3 cells. We also found that ganglioside GM3 interacted with TGFβRs (TGF-β receptors) in TGF-β1-induced HLE B-3 cells. Taken together, these results suggest that ganglioside GM3 induced by TGF-β1 regulates EMT by potential interaction with TGFβRs.
Collapse
|
48
|
Samarakoon R, Overstreet JM, Higgins PJ. TGF-β signaling in tissue fibrosis: redox controls, target genes and therapeutic opportunities. Cell Signal 2012; 25:264-8. [PMID: 23063463 DOI: 10.1016/j.cellsig.2012.10.003] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/07/2012] [Indexed: 10/27/2022]
Abstract
During development of TGF-β1-initiated fibroproliferative disorders, NADPH oxidases (NOX family members) generate reactive oxygen species (ROS) resulting in downstream transcription of a subset genes encoding matrix structural elements and profibrotic factors. Prominent among the repertoire of disease-implicated genes is the TGF-β1 target gene encoding the potent profibrotic matricellular protein plasminogen activator inhibitor-1 (PAI-1 or SERPINE1). PAI-1 is the major physiologic inhibitor of the plasmin-based pericellular cascade and a causative factor in the development of vascular thrombotic and fibroproliferative disorders. ROS generation in response to TGF-β1 stimulation is rapid and precedes PAI-1 induction; engagement of non-SMAD (e.g., EGFR, Src kinase, MAP kinases, p53) and SMAD2/3 pathways are both required for PAI-1 expression and are ROS-dependent. Recent findings suggest a novel role for p53 in TGF-β1-induced PAI-1 transcription that involves ROS generation and p53/SMAD interactions. Targeting ROS and ROS-activated cellular events is likely to have therapeutic implications in the management of fibrotic disorders, particularly in the context of prolonged TGF-β1 signaling.
Collapse
Affiliation(s)
- Rohan Samarakoon
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | |
Collapse
|
49
|
Zfp423 promotes adipogenic differentiation of bovine stromal vascular cells. PLoS One 2012; 7:e47496. [PMID: 23071815 PMCID: PMC3468566 DOI: 10.1371/journal.pone.0047496] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 09/17/2012] [Indexed: 01/09/2023] Open
Abstract
Intramuscular fat or marbling is critical for the palatability of beef. In mice, very recent studies show that adipocytes and fibroblasts share a common pool of progenitor cells, with Zinc finger protein 423 (Zfp423) as a key initiator of adipogenic differentiation. To evaluate the role of Zfp423 in intramuscular adipogenesis and marbling in beef cattle, we sampled beef muscle for separation of stromal vascular cells. These cells were immortalized with pCI neo-hEST2 and individual clones were selected by G418. A total of 288 clones (3×96 well plates) were isolated and induced to adipogenesis. The presence of adipocytes was assessed by Oil-Red-O staining. Three clones with high and low adipogenic potential respectively were selected for further analyses. In addition, fibro/adipogenic progenitor cells were selected using a surface marker, platelet derived growth factor receptor (PDGFR) α. The expression of Zfp423 was much higher (307.4±61.9%, P<0.05) in high adipogenic cells, while transforming growth factor (TGF)-β was higher (156.1±48.7%, P<0.05) in low adipogenic cells. Following adipogenic differentiation, the expression of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα) were much higher (239.4±84.1% and 310.7±138.4%, respectively, P<0.05) in high adipogenic cells. Over-expression of Zfp423 in stromal vascular cells and cloned low adipogenic cells dramatically increased their adipogenic differentiation, accompanied with the inhibition of TGF-β expression. Zfp423 knockdown by shRNA in high adipogenic cells largely prevented their adipogenic differentiation. The differential regulation of Zfp423 and TGF-β between low and high adipogenic cells is associated with the DNA methylation in their promoters. In conclusion, data show that Zfp423 is a critical regulator of adipogenesis in stromal vascular cells of bovine muscle, and Zfp423 may provide a molecular target for enhancing intramuscular adipogenesis and marbling in beef cattle.
Collapse
|
50
|
Reid CD, Zhang Y, Sheets MD, Kessler DS. Transcriptional integration of Wnt and Nodal pathways in establishment of the Spemann organizer. Dev Biol 2012; 368:231-41. [PMID: 22627292 DOI: 10.1016/j.ydbio.2012.05.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/22/2012] [Accepted: 05/08/2012] [Indexed: 11/25/2022]
Abstract
Signaling inputs from multiple pathways are essential for the establishment of distinct cell and tissue types in the embryo. Therefore, multiple signals must be integrated to activate gene expression and confer cell fate, but little is known about how this occurs at the level of target gene promoters. During early embryogenesis, Wnt and Nodal signals are required for formation of the Spemann organizer, which is essential for germ layer patterning and axis formation. Signaling by both Wnt and Nodal pathways is required for the expression of multiple organizer genes, suggesting that integration of these signals is required for organizer formation. Here, we demonstrate transcriptional cooperation between the Wnt and Nodal pathways in the activation of the organizer genes Goosecoid (Gsc), Cerberus (Cer), and Chordin (Chd). Combined Wnt and Nodal signaling synergistically activates transcription of these organizer genes. Effectors of both pathways occupy the Gsc, Cer and Chd promoters and effector occupancy is enhanced with active Wnt and Nodal signaling. This suggests that, at organizer gene promoters, a stable transcriptional complex containing effectors of both pathways forms in response to combined Wnt and Nodal signaling. Consistent with this idea, the histone acetyltransferase p300 is recruited to organizer promoters in a Wnt and Nodal effector-dependent manner. Taken together, these results offer a mechanism for spatial and temporal restriction of organizer gene transcription by the integration of two major signaling pathways, thus establishing the Spemann organizer domain.
Collapse
Affiliation(s)
- Christine D Reid
- Department of Cell and Developmental Biology, University of Pennsylvania, School of Medicine, Room 1110 Biomedical Research Building 2/3, 421 Curie Boulevard, Philadelphia, PA 19104-6058, USA
| | | | | | | |
Collapse
|