1
|
Jung SC, Kang D, Ko EA. Roles of PDGF/PDGFR signaling in various organs. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:139-155. [PMID: 39482238 PMCID: PMC11842291 DOI: 10.4196/kjpp.24.309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Platelet-derived growth factors (PDGFs) ligands and their corresponding receptors, PDGF receptor (PDGFR)α and PDGFRβ, play a crucial role in controlling diverse biological functions, including cell growth, viability and migration. These growth factors bind to PDGFRs, which are receptor tyrosine kinases present on the surface of target cells. The interaction between PDGFs and PDGFRs induces receptor dimerization and subsequent activation through auto-phosphorylation, which in turn triggers a cascade of intracellular signaling pathways. PDGF/PDGFR signaling is essential for maintaining normal physiological functions, including tissue regeneration and growth. However, dysregulation of this signaling pathway leads to pathological conditions, including fibrosis, atherosclerosis, and cancer development in various organs. The pathological impact of PDGF/PDGFR signaling primarily stems from its capacity to promote excessive cell proliferation, enhanced migration, and increased extracellular matrix deposition, resulting in tissue overgrowth, scarring, and abnormal vessel formation. These processes are integral to the pathogenesis of fibrotic, neoplastic, and vascular disorders. Therefore, understanding these pathways is crucial for developing targeted treatments designed to inhibit PDGF/PDGFR signaling in these diseases. This review delves into the dual role of PDGF/PDGFR signaling in both physiological and pathophysiological contexts across different organs and provides insights into current pharmacological therapies designed to target the PDGF signaling pathway.
Collapse
Affiliation(s)
- Sung-Cherl Jung
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| | - Dawon Kang
- Department of Physiology, College of Medicine and Institute of Medical Sciences, Gyeongsang National University, Jinju 52727, Korea
| | - Eun-A Ko
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
2
|
Moreno J, Gluud LL, Galsgaard ED, Hvid H, Mazzoni G, Das V. Identification of ligand and receptor interactions in CKD and MASH through the integration of single cell and spatial transcriptomics. PLoS One 2024; 19:e0302853. [PMID: 38768139 PMCID: PMC11104622 DOI: 10.1371/journal.pone.0302853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Chronic Kidney Disease (CKD) and Metabolic dysfunction-associated steatohepatitis (MASH) are metabolic fibroinflammatory diseases. Combining single-cell (scRNAseq) and spatial transcriptomics (ST) could give unprecedented molecular disease understanding at single-cell resolution. A more comprehensive analysis of the cell-specific ligand-receptor (L-R) interactions could provide pivotal information about signaling pathways in CKD and MASH. To achieve this, we created an integrative analysis framework in CKD and MASH from two available human cohorts. RESULTS The analytical framework identified L-R pairs involved in cellular crosstalk in CKD and MASH. Interactions between cell types identified using scRNAseq data were validated by checking the spatial co-presence using the ST data and the co-expression of the communicating targets. Multiple L-R protein pairs identified are known key players in CKD and MASH, while others are novel potential targets previously observed only in animal models. CONCLUSION Our study highlights the importance of integrating different modalities of transcriptomic data for a better understanding of the molecular mechanisms. The combination of single-cell resolution from scRNAseq data, combined with tissue slide investigations and visualization of cell-cell interactions obtained through ST, paves the way for the identification of future potential therapeutic targets and developing effective therapies.
Collapse
Affiliation(s)
- Jaime Moreno
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Henning Hvid
- Global Drug Discovery, Novo Nordisk A/S, Maløv, Denmark
| | - Gianluca Mazzoni
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| | - Vivek Das
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| |
Collapse
|
3
|
Yoo SH, Nahm JH, Lee WK, Lee HW, Chang HY, Lee JI. Loss of Krüppel-like factor-10 facilitates the development of chemical-induced liver cancer in mice. Mol Med 2023; 29:156. [PMID: 37946098 PMCID: PMC10636809 DOI: 10.1186/s10020-023-00751-1] [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: 05/10/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Krüppel-like factor 10 (KLF10) is involved in a positive feedback loop that regulates transforming growth factor β (TGFβ) signaling, and TGFβ plays an important role in the pathogenesis of liver disease. Here, we investigated whether KLF10 deletion affects the development of liver fibrosis and hepatocellular carcinoma (HCC). METHODS We induced KLF10 deletion in C57BL/6 mice. Liver fibrosis was induced by feeding a diet high in fat and sucrose (high-fat diet [HFD]), whereas HCC was produced by intraperitoneal administration of N-diethylnitrosamine (DEN). An in vitro experiment was performed to evaluate the role of KLF10 in the cancer microenvironment using Hep3B and LX2 cells. An immunohistochemical study of KLF10 expression was performed using human HCC samples from 60 patients who had undergone liver resection. RESULTS KLF10 deletion resulted in an increased DEN-induced HCC burden with significant upregulation of SMAD2, although loss of KLF10 did not alter HFD-induced liver fibrosis. DEN-treated mice with KLF10 deletion exhibited increased levels of mesenchymal markers (N-cadherin and SNAI2) and tumor metastasis markers (matrix metalloproteinases 2 and 9). KLF10 depletion in Hep3B and LX2 cells using siRNA was associated with increased invasiveness. Compared with co-culture of KLF10-preserved Hep3B cells and KLF10-intact LX2 cells, co-culture of KLF10-preserved Hep3B cells and KLF10-depleted LX2 cells resulted in significantly enhanced invasion. Low KLF10 expression in resected human HCC specimens was associated with poor survival. CONCLUSION The results of this study suggest that loss of KLF10 facilitates liver cancer development with alteration in TGFβ signaling.
Collapse
Affiliation(s)
- Sung Hwan Yoo
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Ji Hae Nahm
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Woon Kyu Lee
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Hyun Woong Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea
| | - Hye Young Chang
- Medical Research Center, Gangnam Severance Hospital, Seoul, 06230, Republic of Korea
| | - Jung Il Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
4
|
Enomoto Y, Katsura H, Fujimura T, Ogata A, Baba S, Yamaoka A, Kihara M, Abe T, Nishimura O, Kadota M, Hazama D, Tanaka Y, Maniwa Y, Nagano T, Morimoto M. Autocrine TGF-β-positive feedback in profibrotic AT2-lineage cells plays a crucial role in non-inflammatory lung fibrogenesis. Nat Commun 2023; 14:4956. [PMID: 37653024 PMCID: PMC10471635 DOI: 10.1038/s41467-023-40617-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
The molecular etiology of idiopathic pulmonary fibrosis (IPF) has been extensively investigated to identify new therapeutic targets. Although anti-inflammatory treatments are not effective for patients with IPF, damaged alveolar epithelial cells play a critical role in lung fibrogenesis. Here, we establish an organoid-based lung fibrosis model using mouse and human lung tissues to assess the direct communication between damaged alveolar type II (AT2)-lineage cells and lung fibroblasts by excluding immune cells. Using this in vitro model and mouse genetics, we demonstrate that bleomycin causes DNA damage and activates p53 signaling in AT2-lineage cells, leading to AT2-to-AT1 transition-like state with a senescence-associated secretory phenotype (SASP). Among SASP-related factors, TGF-β plays an exclusive role in promoting lung fibroblast-to-myofibroblast differentiation. Moreover, the autocrine TGF-β-positive feedback loop in AT2-lineage cells is a critical cellular system in non-inflammatory lung fibrogenesis. These findings provide insights into the mechanism of IPF and potential therapeutic targets.
Collapse
Affiliation(s)
- Yasunori Enomoto
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Hiroaki Katsura
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Takashi Fujimura
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Department of Drug Modality Development, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 5-1-35 Saitoaokita, Minoh, 562-0029, Japan
| | - Akira Ogata
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Saori Baba
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Akira Yamaoka
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Miho Kihara
- Laboratory for Animal Resources and Genetic Engineering (LARGE), RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering (LARGE), RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Osamu Nishimura
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Mitsutaka Kadota
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Daisuke Hazama
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yugo Tanaka
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshimasa Maniwa
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Mitsuru Morimoto
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
5
|
Beaven E, Kumar R, Bhatt HN, Esquivel SV, Nurunnabi M. Myofibroblast specific targeting approaches to improve fibrosis treatment. Chem Commun (Camb) 2022; 58:13556-13571. [PMID: 36445310 PMCID: PMC9946855 DOI: 10.1039/d2cc04825f] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Fibrosis has been shown to develop in individuals with underlying health conditions, especially chronic inflammatory diseases. Fibrosis is often diagnosed in various organs, including the liver, lungs, kidneys, heart, and skin, and has been described as excessive accumulation of extracellular matrix that can affect specific organs in the body or systemically throughout the body. Fibrosis as a chronic condition can result in organ failure and result in death of the individual. Understanding and identification of specific biomarkers associated with fibrosis has emerging potential in the development of diagnosis and targeting treatment modalities. Therefore, in this review, we will discuss multiple signaling pathways such as TGF-β, collagen, angiotensin, and cadherin and outline the chemical nature of the different signaling pathways involved in fibrogenesis as well as the mechanisms. Although it has been well established that TGF-β is the main catalyst initiating and driving multiple pathways for fibrosis, targeting TGF-β can be challenging as this molecule regulates essential functions throughout the body that help to keep the body in homeostasis. We also discuss collagen, angiotensin, and cadherins and their role in fibrosis. We comprehensively discuss the various delivery systems used to target collagen, angiotensin, and cadherins to manage fibrosis. Nevertheless, understanding the steps by which this molecule drives fibrosis development can aid in the development of specific targets of its cascading mechanism. Throughout the review, we will demonstrate the mechanism of fibrosis targeting to improve targeting delivery and therapy.
Collapse
Affiliation(s)
- Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA.
- Department of Biomedical Engineering, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA.
- Department of Biomedical Engineering, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA.
- Department of Biomedical Engineering, The University of Texas El Paso, El Paso, TX 79968, USA
| | - Stephanie V Esquivel
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA.
- Aerospace Center (cSETR), The University of Texas El Paso, El Paso, TX 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA.
- Department of Biomedical Engineering, The University of Texas El Paso, El Paso, TX 79968, USA
- Aerospace Center (cSETR), The University of Texas El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, USA
| |
Collapse
|
6
|
Tian Y, Zhan Y, Jiang Q, Lu W, Li X. Expression and function of PDGF-C in development and stem cells. Open Biol 2021; 11:210268. [PMID: 34847773 PMCID: PMC8633783 DOI: 10.1098/rsob.210268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Platelet-derived growth factor C (PDGF-C) is a relatively new member of the PDGF family, discovered nearly 20 years after the finding of platelet-derived growth factor A (PDGF-A) and platelet-derived growth factor B (PDGF-B). PDGF-C is generally expressed in most organs and cell types. Studies from the past 20 years have demonstrated critical roles of PDGF-C in numerous biological, physiological and pathological processes, such as development, angiogenesis, tumour growth, tissue remodelling, wound healing, atherosclerosis, fibrosis, stem/progenitor cell regulation and metabolism. Understanding PDGF-C expression and activities thus will be of great importance to various research disciplines. In this review, however, we mainly discuss the expression and functions of PDGF-C and its receptors in development and stem cells.
Collapse
Affiliation(s)
- Yi Tian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| | - Ying Zhan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| | - Qin Jiang
- Ophthalmic Department, Affiliated Eye Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Weisi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| |
Collapse
|
7
|
Crosstalk between hepatic stellate cells and tumor cells in the development of hepatocellular carcinoma. Chin Med J (Engl) 2021; 134:2544-2546. [PMID: 34520418 PMCID: PMC8577661 DOI: 10.1097/cm9.0000000000001726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
8
|
Qi P, Ma MZ, Kuai JH. Identification of growth differentiation factor 15 as a pro-fibrotic factor in mouse liver fibrosis progression. Int J Exp Pathol 2021; 102:148-156. [PMID: 33983642 DOI: 10.1111/iep.12398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/08/2021] [Accepted: 03/21/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was elucidate the inhibitory role of growth differentiation factor 15 (GDF15) in liver fibrosis and its possible activation mechanism in hepatic stellate cells (HSCs) of mice. We generated a GDF15-neutralizing antibody that can inhibit TGF-β1-induced activation of the TGF-β/Smad2/3 pathway in LX-2 cells. All the mice in this study were induced by carbon tetrachloride and thioacetamide. In addition, primary HSCs from mice were isolated from fresh livers using Nycodenz density gradient separation. The severity and extent of liver fibrosis were evaluated by Sirius Red and Masson staining. The effect of GDF15 on the activation of the TGF-β pathway was detected using dual-luciferase reporter and Western blotting assays. The expression of GDF15 in cirrhotic liver tissue was higher than that in normal liver tissue. Blocking GDF15 with a neutralizing antibody resulted in a delay in primary hepatic stellate cell activation and remission of liver fibrosis induced by carbon tetrachloride or thioacetamide. Meanwhile, TGF-β pathway activation was partly inhibited by a GDF15-neutralizing antibody in primary HSCs. These results indicated that GDF15 plays an important role in regulating HSC activation and liver fibrosis progression. The inhibition of GDF15 attenuates chemical-inducible liver fibrosis and delays hepatic stellate cell activation, and this effect is probably mainly attributed to its regulatory role in TGF-β signalling.
Collapse
Affiliation(s)
- Peng Qi
- Department of Cardiac Surgery Intensive Care Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Ming-Ze Ma
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing-Hua Kuai
- Department of Gastroenterology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
9
|
Wang L, Chang M, Tian Y, Yan J, Xu W, Yuan S, Zhang K, Liu X. The Role of Smad2 in Transforming Growth Factor β 1-Induced Hypertrophy of Ligamentum Flavum. World Neurosurg 2021; 151:e128-e136. [PMID: 33831616 DOI: 10.1016/j.wneu.2021.03.147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/29/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Hypertrophy of the ligamentum flavum (LF) contributes to the development of spinal stenosis. Smad proteins can mediate the fibrogenesis activity through the transforming growth factor β1 (TGF-β1) pathway, but which Smad protein plays a more important role in the hypertrophy process of LF is unclear. METHODS The LF samples were obtained from 50 patients. After the LF cells (LFCs) were cultured, small interfering ribonucleic acid (siRNA) that target human phosphorylated-Smad2, 3, or 4 (p-Smad2,3,4) genes was transfected into LFCs. Next, proteins from cells were extracted and the protein levels of Smad2, Smad3, and Smad4 were detected by Western blot. The messenger ribonucleic acid level of TGF-β1 was measured by real-time polymerase chain reaction (PCR). Furthermore, an enzyme-linked immunosorbent assay was performed to test the impact of Smad2 downstream of the TGF-β1 signaling pathway. RESULTS Degeneration of the LF was characterized by an increase in disorganized elastic fibers and fibrotic transformation by extracellular collagen deposition. The gene expression analysis of fibrotic genes in LFCs showed that knockdown of phosphorylated-Smad2 by siRNA significantly reduced the protein expression level of TGF-β1 compared with other groups. The enzyme-linked immunosorbent assay suggested that the protein expression level of Smad2 can influence the downstream events of TGF-β1 signaling pathway in the LFCs. CONCLUSIONS Our findings suggest that Smad2 plays a potential role in the pathologic development of hypertrophy of LF. We also found that Smad2 knockdown by Smad-siRNA can influence the TGF-β1 signaling pathway through decreasing expression of TGF-β1, tumor necrosis factor α, and nuclear factor κb.
Collapse
Affiliation(s)
- Lianlei Wang
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China
| | - Mingzheng Chang
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P. R. China
| | - Yonghao Tian
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China
| | - Jun Yan
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China
| | - Wanlong Xu
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China
| | - Suomao Yuan
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China
| | - Kai Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China
| | - Xinyu Liu
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P. R. China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P. R. China.
| |
Collapse
|
10
|
Sharawy MH, El-Awady MS, Makled MN. Protective effects of paclitaxel on thioacetamide-induced liver fibrosis in a rat model. J Biochem Mol Toxicol 2021; 35:e22745. [PMID: 33749060 DOI: 10.1002/jbt.22745] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 01/18/2023]
Abstract
Liver fibrosis is a public health burden that is highly associated with morbidity and mortality. Therefore, this study aims to explore the anti-fibrotic effects of low dose of paclitaxel (PTX) against thioacetamide (TAA)-induced liver fibrosis in rats and the possible mechanisms involved. TAA was administered at a dose of 200 mg/kg twice weekly for 6 weeks in rats to induce liver fibrosis similar to that in humans. Liver dysfunction was shown by increased alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyl transferase, along with histopathological changes. Liver fibrosis was confirmed by Masson's Trichome staining, increased collagen content, and elevated α-smooth muscle actin (α-SMA) protein expression. In addition, TAA induced liver apoptosis as indicated by the increased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in liver tissues. This study demonstrated that the administration of PTX (0.3 mg/kg/i.p.) three times a week for 6 weeks significantly alleviated functional and biochemical changes induced by TAA in addition to improving the liver architecture. PTX attenuated liver fibrosis as reflected by the decreased collagen content and α-SMA protein expression. Additionally, PTX attenuated liver apoptosis as indicated by the decreased TUNEL-positive cells. Moreover, PTX prevented TAA-induced elevation of transforming growth factor-β1 (TGF-β1), platelet-derived growth factor-BB (PDGF-BB), and tissue inhibitor of metalloproteinase 1 (TIMP-1) levels in liver tissues. These findings suggest that the low dose of PTX prevented TAA-induced liver fibrosis in rats, possibly by inhibiting the expression of TGF-β1 and PDGF-BB and subsequently suppressing the apoptosis and the expression of TIMP-1.
Collapse
Affiliation(s)
- Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohammed S El-Awady
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.,Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
11
|
Heyens LJM, Busschots D, Koek GH, Robaeys G, Francque S. Liver Fibrosis in Non-alcoholic Fatty Liver Disease: From Liver Biopsy to Non-invasive Biomarkers in Diagnosis and Treatment. Front Med (Lausanne) 2021; 8:615978. [PMID: 33937277 PMCID: PMC8079659 DOI: 10.3389/fmed.2021.615978] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
An increasing percentage of people have or are at risk to develop non-alcoholic fatty liver disease (NAFLD) worldwide. NAFLD comprises different stadia going from isolated steatosis to non-alcoholic steatohepatitis (NASH). NASH is a chronic state of liver inflammation that leads to the transformation of hepatic stellate cells to myofibroblasts. These cells produce extra-cellular matrix that results in liver fibrosis. In a normal situation, fibrogenesis is a wound healing process that preserves tissue integrity. However, sustained and progressive fibrosis can become pathogenic. This process takes many years and is often asymptomatic. Therefore, patients usually present themselves with end-stage liver disease e.g., liver cirrhosis, decompensated liver disease or even hepatocellular carcinoma. Fibrosis has also been identified as the most important predictor of prognosis in patients with NAFLD. Currently, only a minority of patients with liver fibrosis are identified to be at risk and hence referred for treatment. This is not only because the disease is largely asymptomatic, but also due to the fact that currently liver biopsy is still the golden standard for accurate detection of liver fibrosis. However, performing a liver biopsy harbors some risks and requires resources and expertise, hence is not applicable in every clinical setting and is unsuitable for screening. Consequently, different non-invasive diagnostic tools, mainly based on analysis of blood or other specimens or based on imaging have been developed or are in development. In this review, we will first give an overview of the pathogenic mechanisms of the evolution from isolated steatosis to fibrosis. This serves as the basis for the subsequent discussion of the current and future diagnostic biomarkers and anti-fibrotic drugs.
Collapse
Affiliation(s)
- Leen J. M. Heyens
- Faculty of Health and Life Sciences, Hasselt University, Hasselt, Belgium
- School of Nutrition and Translational Research in Metabolism, NUTRIM, Maastricht University, Maastricht, Netherlands
- Department of Gastro-Enterology and Hepatology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Dana Busschots
- Faculty of Health and Life Sciences, Hasselt University, Hasselt, Belgium
- School of Nutrition and Translational Research in Metabolism, NUTRIM, Maastricht University, Maastricht, Netherlands
| | - Ger H. Koek
- School of Nutrition and Translational Research in Metabolism, NUTRIM, Maastricht University, Maastricht, Netherlands
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Geert Robaeys
- Faculty of Health and Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Gastro-Enterology and Hepatology, Ziekenhuis Oost-Limburg, Genk, Belgium
- Department of Gastroenterology and Hepatology, University Hospital Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- *Correspondence: Sven Francque
| |
Collapse
|
12
|
Jiang H, Zhang L, Liu X, Sun W, Kato K, Chen C, Li X, Li T, Sun Z, Han W, Zhang F, Xiao Q, Yang Z, Hu J, Qin Z, Adams RH, Gao X, He Y. Angiocrine FSTL1 (Follistatin-Like Protein 1) Insufficiency Leads to Atrial and Venous Wall Fibrosis via SMAD3 Activation. Arterioscler Thromb Vasc Biol 2020; 40:958-972. [PMID: 32078339 DOI: 10.1161/atvbaha.119.313901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Angiocrine factors, mediating the endothelial-mural cell interaction in vascular wall construction as well as maintenance, are incompletely characterized. This study aims to investigate the role of endothelial cell-derived FSTL1 (follistatin-like protein 1) in vascular homeostasis. Approach and Results: Using conditional knockout mouse models, we show that loss of FSTL1 in endothelial cells (Fstl1ECKO) led to an increase of pulmonary vascular resistance, resulting in the heart regurgitation especially with tricuspid valves. However, this abnormality was not detected in mutant mice with Fstl1 knockout in smooth muscle cells or hematopoietic cells. We further showed that there was excessive αSMA (α-smooth muscle actin) associated with atrial endocardia, heart valves, veins, and microvessels after the endothelial FSTL1 deletion. There was also an increase in collagen deposition, as demonstrated in livers of Fstl1ECKO mutants. The SMAD3 (mothers against decapentaplegic homolog 3) phosphorylation (pSMAD3) was significantly enhanced, and pSMAD3 staining was colocalized with αSMA in vein walls, suggesting the activation of TGFβ (transforming growth factor β) signaling in vascular mural cells of Fstl1ECKO mice. Consistently, treatment with a TGFβ pathway inhibitor reduced the abnormal association of αSMA with the atria and blood vessels in Fstl1ECKO mutant mice. CONCLUSIONS The findings imply that endothelial FSTL1 is critical for the homeostasis of vascular walls, and its insufficiency may favor cardiovascular fibrosis leading to heart failure.
Collapse
Affiliation(s)
- Haijuan Jiang
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Luqing Zhang
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.).,MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Xuelian Liu
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Wei Sun
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.).,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, China (W.S.)
| | - Katsuhiro Kato
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Germany (K.K., R.H.A.).,Department of Cardiology, Nagoya University Hospital, Japan (K.K.)
| | - Chuankai Chen
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Xiao Li
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Taotao Li
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Zhiliang Sun
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Wencan Han
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Fujing Zhang
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Qi Xiao
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Zhongzhou Yang
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, China (J.H.)
| | - Zhihai Qin
- The First Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, China (Z.Q.)
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Germany (K.K., R.H.A.)
| | - Xiang Gao
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Yulong He
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| |
Collapse
|
13
|
Distler JHW, Györfi AH, Ramanujam M, Whitfield ML, Königshoff M, Lafyatis R. Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol 2019; 15:705-730. [PMID: 31712723 DOI: 10.1038/s41584-019-0322-7] [Citation(s) in RCA: 390] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
|
14
|
Karimi A, Shahrooz R, Hobbenagh R, Delirezh N, Amani S, Garssen J, Mortaz E, M Adcock I. Histological Evidence for Therapeutic Induction of Angiogenesis Using Mast Cells and Platelet-Rich Plasma within A Bioengineered Scaffold following Rat Hindlimb Ischemia. CELL JOURNAL 2019; 21:391-400. [PMID: 31376320 PMCID: PMC6722454 DOI: 10.22074/cellj.2020.6287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/26/2018] [Indexed: 11/29/2022]
Abstract
Objective Peripheral arterial disease results from obstructed blood flow in arteries and increases the risk of amputation
in acute cases. Therapeutic angiogenesis using bioengineered tissues composed of a chitosan scaffold that was
enriched with mast cells (MCs) and/or platelet-rich plasma (PRP) was used to assess the formation of vascular networks
and subsequently improved the functional recovery following hindlimb ischemia. This study aimed to find an optimal
approach for restoring local vascularization.
Materials and Methods In this experimental study, thirty rats were randomly divided into six experimental groups: a.
Ischemic control group with right femoral artery transection, b. Ischemia with phosphate-buffered saline (PBS) control
group, c. Ischemia with chitosan scaffold, d. Ischemia with chitosan and MCs, e. Ischemia with chitosan and PRP, and
f. Ischemia with chitosan, PRP, and MCs. The left hind limbs served as non-ischemic controls. The analysis of capillary
density, arterial diameter, histomorphometric analysis and immunohistochemistry at the transected locations and in
gastrocnemius muscles was performed.
Results The group treated with chitosan/MC significantly increased capillary density and the mean number of
large blood vessels at the site of femoral artery transection compared with other experimental groups (P<0.05). The
treatment with chitosan/MC also significantly increased the muscle fiber diameter and the capillary-to-muscle fiber ratio
in gastrocnemius muscles compared with all other ischemic groups (P<0.05).
Conclusion These findings suggested that chitosan and MCs together could offer a new approach for the therapeutic
induction of angiogenesis in cases of peripheral arterial diseases.
Collapse
Affiliation(s)
- Ali Karimi
- Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rasoul Shahrooz
- Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.Electronic Address:
| | - Rahim Hobbenagh
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Nowruz Delirezh
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Saeede Amani
- Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| | - Esmaeil Mortaz
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Tuberculosis and Epidemiology Research Center, National Research Institute for Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.Electronic Address:
| | - Ian M Adcock
- Cell and Molecular Biology Group, Airways Disease Section, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
15
|
Song LY, Ma YT, Fang WJ, He Y, Wu JL, Zuo SR, Deng ZZ, Wang SF, Liu SK. Inhibitory effects of oxymatrine on hepatic stellate cells activation through TGF-β/miR-195/Smad signaling pathway. Altern Ther Health Med 2019; 19:138. [PMID: 31221141 PMCID: PMC6585021 DOI: 10.1186/s12906-019-2560-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/11/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Oxymatrine (OM), a quinolizidine alkaloid extracted from a herb Sophorae Flavescentis Radix, has been used to treat liver fibrotic diseases. However, the mechanism of its anti-fibrosis effects is still unclear. TGF-β/Smad signaling and miR-195 have been proved to paly an important role in hepatic stellate cells (HSCs) activation and liver fibrosis. In this study, we investigated whether OM could inhibit HSCs activation through TGF-β1/miR-195/Smads signaling or not. METHODS First, the effects of OM on HSC-T6 in different concentrations and time points were tested by MTT assay. We choose three appropriate concentrations of OM as treatment concentrations in following experiment. By Quantitative Real-time PCR and Western Blot, then we investigated the effect of OM on miR-195, Smad7 and α-SMA's expressions to prove the correlation between OM and the TGF-β1/miR-195/Smads signaling. Last, miR-195 mimic and INF-γ were used to investigate the relation between miR-195 and OM in HSC activation. RESULTS Our results showed that the proliferation of HSC was significantly inhibited when OM concentration was higher than 200 μg/mL after 24 h, 100 μg/mL after 48 h and 10 μg/mL after 72 h. The IC50 of OM after 24, 48 and 72 h were 539, 454, 387 μg/mL respectively. OM could down-regulate miR-195 and α-SMA (P < 0.01), while up-regulate Smad7 (P < 0.05). In HSC-T6 cells transfected with miR-195 mimic and pretreated with OM, miR-195 and α-SMA were up-regulated (P < 0.05), and Smad7 was down-regulated (P < 0.05) . CONCLUSIONS Given these results, OM could inhibit TGF-β1 induced activation of HSC-T6 proliferation in a dose-dependent and time-dependent manner to some extent. We proved that OM inhibited HSC activation through down-regulating the expression of miR-195 and up-regulating Smad7.
Collapse
|
16
|
Wang L, Liu Q, Kitamoto T, Hou J, Qin J, Accili D. Identification of Insulin-Responsive Transcription Factors That Regulate Glucose Production by Hepatocytes. Diabetes 2019; 68:1156-1167. [PMID: 30936148 PMCID: PMC6610019 DOI: 10.2337/db18-1236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/20/2019] [Indexed: 01/02/2023]
Abstract
Hepatocyte glucose production is a complex process that integrates cell-autonomous mechanisms with cellular signaling, enzyme activity modulation, and gene transcription. Transcriptional mechanisms controlling glucose production are redundant and involve nuclear hormone receptors and unliganded transcription factors (TFs). Our knowledge of this circuitry is incomplete. Here we used DNA affinity purification followed by mass spectrometry to probe the network of hormone-regulated TFs by using phosphoenolpyruvate carboxykinase (Pck1) and glucose-6-phosphatase (G6pc) in liver and primary hepatocytes as model systems. The repertoire of insulin-regulated TFs is unexpectedly broad and diverse. Whereas in liver the two test promoters are regulated by largely overlapping sets of TFs, in primary hepatocytes Pck1 and G6pc regulation diverges. Insulin treatment preferentially results in increased occupancy by the two promoters, consistent with a model in which the hormone's primary role is to recruit corepressors rather than to clear activators. Nine insulin-responsive TFs are present in both models, but only FoxK1, FoxA2, ZFP91, and ZHX3 require an intact Pck1p insulin response sequence for binding. Knockdown of FoxK1 in primary hepatocytes decreased both glucose production and insulin's ability to suppress it. The findings expand the repertoire of insulin-dependent TFs and identify FoxK1 as a contributor to insulin signaling.
Collapse
Affiliation(s)
- Liheng Wang
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Qiongming Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences, Beijing, China
| | - Takumi Kitamoto
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Junjie Hou
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences, Beijing, China
| | - Domenico Accili
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
17
|
Yun SM, Kim SH, Kim EH. The Molecular Mechanism of Transforming Growth Factor-β Signaling for Intestinal Fibrosis: A Mini-Review. Front Pharmacol 2019; 10:162. [PMID: 30873033 PMCID: PMC6400889 DOI: 10.3389/fphar.2019.00162] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Inflammatory bowel disease is known as the most chronic inflammatory disorder in colon, which subsequently progresses to intestinal obstruction and fistula formation. Many studies to date for the treatment of IBD have been focused on inflammation. However, most of the anti-inflammatory agents do not have anti-fibrotic effects and could not relieve intestinal stricture in IBD patients. Because preventing or reversing intestinal fibrosis in IBD is a major therapeutic target, we analyzed the papers focusing on TGF-β signaling in intestinal fibrosis. TGF-β is a good candidate to treat the intestinal fibrosis in IBD which involves TGF-β signaling pathway, EMT, EndMT, ECM, and other regulators. Understanding the mechanism involved in TGF-β signaling will contribute to the treatment and diagnosis of intestinal fibrosis occurring in IBD as well as the understanding of the molecular mechanisms underlying the pathogenesis.
Collapse
Affiliation(s)
- Sun-Mi Yun
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Seok-Ho Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| |
Collapse
|
18
|
Sato F, Otsuka T, Kohsaka A, Le HT, Bhawal UK, Muragaki Y. Smad3 Suppresses Epithelial Cell Migration and Proliferation via the Clock Gene Dec1, Which Negatively Regulates the Expression of Clock Genes Dec2 and Per1. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:773-783. [PMID: 30664860 DOI: 10.1016/j.ajpath.2019.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/03/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
Abstract
Smad3 has circadian expression; however, whether Smad3 affects the expression of clock genes is poorly understood. Here, we investigated the regulatory mechanisms between Smad3 and the clock genes Dec1, Dec2, and Per1. In Smad3 knockout mice, the amplitude of locomotor activity was decreased, and Dec1 expression was decreased in the suprachiasmatic nucleus, liver, kidney, and tongue compared with control mice. Conversely, Dec2 and Per1 expression was increased compared with that of control mice. In Smad3 knockout mice, immunohistochemical staining revealed that Dec1 expression decreased, whereas Dec2 and Per1 expression increased in the endothelial cells of the kidney and liver. In NIH3T3 cells, Smad3 overexpression increased Dec1 expression, but decreased Dec2 and Per1 expression. In a wound-healing experiment that used Smad3 knockout mice, Dec1 expression decreased in the basal cells of squamous epithelium, promoting wound healing of the mucosa. Finally, the migration and proliferation of Smad3 knockdown squamous carcinoma cells was suppressed by Dec1 overexpression but was promoted by Dec2 overexpression. Dec1 overexpression decreased E-cadherin and proliferating cell nuclear antigen expression, whereas these expression levels were increased by Dec2 overexpression. These results suggest Smad3 is relevant to circadian rhythm and regulates cell migration and proliferation through Dec1, Dec2, and Per1 expression.
Collapse
Affiliation(s)
- Fuyuki Sato
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan.
| | - Tsuyoshi Otsuka
- Department of Physiology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Akira Kohsaka
- Department of Physiology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Hue Thi Le
- Department of Physiology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Ujjal K Bhawal
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Yasuteru Muragaki
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
19
|
Lim BJ, Lee WK, Lee HW, Lee KS, Kim JK, Chang HY, Lee JI. Selective deletion of hepatocyte platelet-derived growth factor receptor α and development of liver fibrosis in mice. Cell Commun Signal 2018; 16:93. [PMID: 30509307 PMCID: PMC6276164 DOI: 10.1186/s12964-018-0306-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/21/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Platelet-derived growth factor receptor α (PDGFRα) expression is increased in activated hepatic stellate cells (HSCs) in cirrhotic liver, while normal hepatocytes express PDGFRα at a negligible level. However, cancerous hepatocytes may show upregulation of PDGFRα, and hepatocellular carcinoma is preceded by chronic liver injury. The role of PDGFRα in non-cancerous hepatocytes and liver fibrosis is unclear. We hypothesized that upon liver injury, PDGFRα in insulted hepatocytes contributes to liver fibrosis by facilitating intercellular crosstalk between hepatocytes and HSCs. METHODS Hepatocytes were isolated from normal and thioacetamide (TAA)-induced cirrhotic livers for assessment of PDGFRα expression. Conditional knock-out (KO) C57BL/6 mice, in which PDGFRα was selectively deleted in hepatocytes, were generated. Liver fibrosis was induced by injecting TAA for 8 weeks. Hep3B cells were transfected with a small interfering RNA (siRNA) (PDGFRα or control) and co-cultured with LX2 cells. RESULTS PDGFRα expression was increased in hepatocytes from fibrotic livers compared to normal livers. Conditional PDGFRα KO mice had attenuated TAA-induced liver fibrosis with decreased HSC activation and proliferation. Immunoblot analyses revealed decreased expression of phospho-p44/42 MAPK in TAA-treated KO mice; these mice also showed almost complete suppression of the upregulation of mouse double minute 2. Although KO mice exhibited increased expression of transforming growth factor (TGF)-β and Smad2/3, this was compensated for by increased expression of inhibitory Smad7. LX2 cells co-cultured with PDGFRα siRNA-infected Hep3B cells showed decreased PDGFRα, α smooth muscle actin, collagen α1(I), TGFβ, and Smad2/3 expression. LX2/PDGFRα-deleted hepatocyte co-culture medium showed decreased PDGF-BB and PDGF-CC levels. CONCLUSIONS Deletion of PDGFRα in hepatocytes attenuated the upregulation of PDGFRα in HSCs after TAA treatment, resulting in decreased liver fibrosis and HSC activation. This suggests that in the event of chronic liver injury, PDGFRα in hepatocytes plays an important role in liver fibrosis by affecting PDGFRα expression in HSCs.
Collapse
Affiliation(s)
- Beom Jin Lim
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woon-Kyu Lee
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Republic of Korea
| | - Hyun Woong Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwan Sik Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ja Kyung Kim
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Young Chang
- Medical Research Center, Gangnam Severance Hospital, Seoul, South Korea
| | - Jung Il Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Kudryashova TV, Shen Y, Pena A, Cronin E, Okorie E, Goncharov DA, Goncharova EA. Inhibitory Antibodies against Activin A and TGF-β Reduce Self-Supported, but Not Soluble Factors-Induced Growth of Human Pulmonary Arterial Vascular Smooth Muscle Cells in Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19102957. [PMID: 30274147 PMCID: PMC6212879 DOI: 10.3390/ijms19102957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Increased growth and proliferation of distal pulmonary artery vascular smooth muscle cells (PAVSMC) is an important pathological component of pulmonary arterial hypertension (PAH). Transforming Growth Factor-β (TGF-β) superfamily plays a critical role in PAH, but relative impacts of self-secreted Activin A, Gremlin1, and TGF-β on PAH PAVSMC growth and proliferation are not studied. Here we report that hyper-proliferative human PAH PAVSMC have elevated secretion of TGF-β1 and, to a lesser extent, Activin A, but not Gremlin 1, and significantly reduced Ser465/467-Smad2 and Ser423/425-Smad3 phosphorylation compared to controls. Media, conditioned by PAH PAVSMC, markedly increased Ser465/467-Smad2, Ser423/425-Smad3, and Ser463/465-Smad1/5 phosphorylation, up-regulated Akt, ERK1/2, and p38 MAPK, and induced significant proliferation of non-diseased PAVSMC. Inhibitory anti-Activin A antibody reduced PAH PAVSMC growth without affecting canonical (Smads) or non-canonical (Akt, ERK1/2, p38 MAPK) effectors. Inhibitory anti-TGF-β antibody significantly reduced P-Smad3, P-ERK1/2 and proliferation of PAH PAVSMC, while anti-Gremlin 1 had no anti-proliferative effect. PDGF-BB diminished inhibitory effects of anti-Activin A and anti-TGF-β antibodies. None of the antibodies affected growth and proliferation of non-diseased PAVSMC induced by PAH PAVSMC-secreted factors. Together, these data demonstrate that human PAH PAVSMC have secretory, proliferative phenotype that could be targeted by anti-Activin A and anti-TGF-β antibodies; potential cross-talk with PDGF-BB should be considered while developing therapeutic interventions.
Collapse
Affiliation(s)
- Tatiana V Kudryashova
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Yuanjun Shen
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Andressa Pena
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Emily Cronin
- Division of Mathematics and Sciences, Walsh University, North Canton, OH 44720, USA.
| | - Evelyn Okorie
- Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Dmitry A Goncharov
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Elena A Goncharova
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
- Division of Pulmonary, Allergy and Critical Care, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15213, USA.
- University of Pittsburgh Department of Bioengineering, Pittsburgh, PA 15213, USA.
| |
Collapse
|
21
|
Fleischer LM, Somaiya RD, Miller GM. Review and Meta-Analyses of TAAR1 Expression in the Immune System and Cancers. Front Pharmacol 2018; 9:683. [PMID: 29997511 PMCID: PMC6029583 DOI: 10.3389/fphar.2018.00683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/06/2018] [Indexed: 12/29/2022] Open
Abstract
Since its discovery in 2001, the major focus of TAAR1 research has been on its role in monoaminergic regulation, drug-induced reward and psychiatric conditions. More recently, TAAR1 expression and functionality in immune system regulation and immune cell activation has become a topic of emerging interest. Here, we review the immunologically-relevant TAAR1 literature and incorporate open-source expression and cancer survival data meta-analyses. We provide strong evidence for TAAR1 expression in the immune system and cancers revealed through NCBI GEO datamining and discuss its regulation in a spectrum of immune cell types as well as in numerous cancers. We discuss connections and logical directions for further study of TAAR1 in immunological function, and its potential role as a mediator or modulator of immune dysregulation, immunological effects of psychostimulant drugs of abuse, and cancer progression.
Collapse
Affiliation(s)
- Lisa M Fleischer
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Rachana D Somaiya
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.,Department of Chemical Engineering, Northeastern University, Boston, MA, United States.,Center for Drug Discovery, Northeastern University, Boston, MA, United States
| |
Collapse
|
22
|
Zhang XH, Chen Y, Li B, Liu JY, Yang CM, Ma MZ. Blocking follistatin-like 1 attenuates liver fibrosis in mice by regulating transforming growth factor-beta signaling. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1112-1122. [PMID: 31938206 PMCID: PMC6958153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/02/2017] [Indexed: 06/10/2023]
Abstract
AIM To elucidate the effect of inhibiting follistatin-like 1 on liver fibrosis and activation of hepatic stellate cells in mice. METHODS We generated a follistatin-like 1 neutralizing antibody that can inhibit TGF-β 1-induced expression of collagen1α1 in primary mouse liver fibroblasts. All of the mice in our study were induced with carbon tetrachloride and thioacetamide. In addition, primary hepatic stellate cells from mice were isolated from fresh livers using density gradient separation. The degree and extent of fibrosis in mouse livers from the different groups were evaluated by Sirius Red and Masson staining. The effect of the follistatin-like 1 neutralizing antibody on proliferation and migration of hepatic stellate cells was detected using CCK-8 and Transwell assays, respectively. RESULTS Expression of follistatin-like 1 in human cirrhotic liver tissue was higher than that in normal liver tissue. Blocking follistatin-like 1 resulted in a delay of primary hepatic stellate cell activation and down-regulation of the migratory capacity of hepatic stellate cells. Blocking follistatin-like 1 also down-regulated TGF-beta signaling in primary hepatic stellate cells from mice. Finally, inhibition of follistatin-like 1 attenuated liver fibrosis and liver function damage in vivo. CONCLUSIONS Inhibiting follistatin-like 1 attenuates liver fibrosis and causes a delay in hepatic stellate cell activation. The effect of follistatin-like 1 on liver fibrosis is mainly attributed to its role in regulating TGF-beta signaling.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Yong Chen
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Bin Li
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Ji-Yong Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Chong-Mei Yang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Ming-Ze Ma
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| |
Collapse
|
23
|
Shen M, Cao J, Shi H. Effects of Estrogen and Estrogen Receptors on Transcriptomes of HepG2 Cells: A Preliminary Study Using RNA Sequencing. Int J Endocrinol 2018; 2018:5789127. [PMID: 30510575 PMCID: PMC6230429 DOI: 10.1155/2018/5789127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/12/2018] [Indexed: 12/23/2022] Open
Abstract
Men have a much higher incidence of hepatocellular carcinoma (HCC), the predominant form of liver cancer, than women, suggesting that estrogens play a protective role in liver cancer development and progression. To begin to understand the potential mechanisms of estrogens' inhibitory effects on HCC development, RNA sequencing was used to generate comprehensive global transcriptome profiles of the human HCC-derived HepG2 cell line following treatment of vehicle (control), estradiol (E2), estrogen receptor alpha- (ERα-) specific agonist 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT), or ERβ-specific agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) using a small set of cells. Gene ontology (GO) analysis identified increased expression of genes involved in the biological process (BP) of response to different stimuli and metabolic processes by E2 and ER agonists, which enhanced molecular function (MF) in various enzyme activities and chemical bindings. Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathway analysis indicated enhanced pathways associated with carbohydrate metabolism, complement and coagulation cascades, and HIF-1 signaling pathway by E2 and ER agonists. GO analysis also identified decreased expression of genes by E2, PPT, and DPN involved in BP related to the cell cycle and cell division, which reduced MF in activity of multiple enzymes and microtubule activity. KEGG analysis indicated that E2, PPT, and DPN suppressed pathways associated with the cell cycle; E2 and PPT suppressed pathways associated with chemical carcinogenesis and drug metabolism, and DPN suppressed DNA replication, recombination, and repair. Collectively, these differentially expressed genes across HepG2 cell transcriptome involving cellular and metabolic processes by E2 and ER agonists provided mechanistic insight into protective effects of estrogens in HCC development.
Collapse
Affiliation(s)
- Minqian Shen
- Department of Biology, Miami University, 700 E. High St., Oxford, OH, USA
| | - Jingyi Cao
- Department of Biology, Miami University, 700 E. High St., Oxford, OH, USA
| | - Haifei Shi
- Department of Biology, Miami University, 700 E. High St., Oxford, OH, USA
| |
Collapse
|
24
|
Saito Y, Chikenji T, Ozasa Y, Fujimiya M, Yamashita T, Gingery A, Iba K. PDGFR Signaling Mediates Hyperproliferation and Fibrotic Responses of Subsynovial Connective Tissue Cells in Idiopathic Carpal Tunnel Syndrome. Sci Rep 2017; 7:16192. [PMID: 29170419 PMCID: PMC5700922 DOI: 10.1038/s41598-017-16443-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022] Open
Abstract
Fibrosis of the subsynovial connective tissue (SSCT) is a pathognomonic change in carpal tunnel syndrome (CTS). Identification of molecular targets and anti-fibrotic therapies could provide new treatment strategies for CTS. The contribution of SSCT cells to fibrosis and the signaling pathways that initiate and aggravate fibrosis in CTS remain unknown. Here we report that platelet-derived growth factor receptor alpha (PDGFRα) positive ( + ) cells accumulate in CTS SSCT and that the presence of fibrotic growth factor, PDGF-AA, results in increased proliferation of PDGFRα+ cells via PI3K/Akt signaling pathway. Although PI3K inhibition decreased proliferation, there was no change in fibrosis-related gene expression. Indeed, protein levels of fibrosis signaling mediator TGF-β remained the same and the second messenger, Smad2/3, accumulated in the nucleus. In contrast AMP-activated protein kinase (AMPK) activation, which can be induced with metformin and AICAR inhibited proliferation, TGF-β expression, and altered cell morphology in SSCT cells. Further we show that AMPK activation by metformin reduced collagen III levels and the ratio of Collagen I to Collagen III. Both AICAR and metformin reduced F-actin and significantly reduced the fiber cross alignment. Our results suggest that PDGFRa signaling may be an important fibrosis target and that activators of AMPK, may be an important therapeutic approach for treating CTS.
Collapse
Affiliation(s)
- Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Yasuhiro Ozasa
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Anne Gingery
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, United States
| | - Kousuke Iba
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
25
|
Branched-chain amino acids prevent hepatic fibrosis and development of hepatocellular carcinoma in a non-alcoholic steatohepatitis mouse model. Oncotarget 2017; 8:18191-18205. [PMID: 28212548 PMCID: PMC5392319 DOI: 10.18632/oncotarget.15304] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 12/27/2016] [Indexed: 01/06/2023] Open
Abstract
Oral supplementation with branched-chain amino acids (BCAA; leucine, isoleucine, and valine) in patients with liver cirrhosis potentially suppresses the incidence of hepatocellular carcinoma (HCC) and improves event-free survival. However, the detailed mechanisms of BCAA action have not been fully elucidated. BCAA were administered to atherogenic and high-fat (Ath+HF) diet-induced nonalcoholic steatohepatitis (NASH) model mice. Liver histology, tumor incidence, and gene expression profiles were evaluated. Ath+HF diet mice developed hepatic tumors at a high frequency at 68 weeks. BCAA supplementation significantly improved hepatic steatosis, inflammation, fibrosis, and tumors in Ath+HF mice at 68 weeks. GeneChip analysis demonstrated the significant resolution of pro-fibrotic gene expression by BCAA supplementation. The anti-fibrotic effect of BCAA was confirmed further using platelet-derived growth factor C transgenic mice, which develop hepatic fibrosis and tumors. In vitro, BCAA restored the transforming growth factor (TGF)-β1-stimulated expression of pro-fibrotic genes in hepatic stellate cells (HSC). In hepatocytes, BCAA restored TGF-β1-induced apoptosis, lipogenesis, and Wnt/β-Catenin signaling, and inhibited the transformation of WB-F344 rat liver epithelial stem-like cells. BCAA repressed the promoter activity of TGFβ1R1 by inhibiting the expression of the transcription factor NFY and histone acetyltransferase p300. Interestingly, the inhibitory effect of BCAA on TGF-β1 signaling was mTORC1 activity-dependent, suggesting the presence of negative feedback regulation from mTORC1 to TGF-β1 signaling. Thus, BCAA induce an anti-fibrotic effect in HSC, prevent apoptosis in hepatocytes, and decrease the incidence of HCC; therefore, BCAA supplementation would be beneficial for patients with advanced liver fibrosis with a high risk of HCC.
Collapse
|
26
|
Lefebvre P, Lalloyer F, Baugé E, Pawlak M, Gheeraert C, Dehondt H, Vanhoutte J, Woitrain E, Hennuyer N, Mazuy C, Bobowski-Gérard M, Zummo FP, Derudas B, Driessen A, Hubens G, Vonghia L, Kwanten WJ, Michielsen P, Vanwolleghem T, Eeckhoute J, Verrijken A, Van Gaal L, Francque S, Staels B. Interspecies NASH disease activity whole-genome profiling identifies a fibrogenic role of PPARα-regulated dermatopontin. JCI Insight 2017; 2:92264. [PMID: 28679947 DOI: 10.1172/jci.insight.92264] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/19/2017] [Indexed: 12/21/2022] Open
Abstract
Nonalcoholic fatty liver disease prevalence is soaring with the obesity pandemic, but the pathogenic mechanisms leading to the progression toward active nonalcoholic steatohepatitis (NASH) and fibrosis, major causes of liver-related death, are poorly defined. To identify key components during the progression toward NASH and fibrosis, we investigated the liver transcriptome in a human cohort of NASH patients. The transition from histologically proven fatty liver to NASH and fibrosis was characterized by gene expression patterns that successively reflected altered functions in metabolism, inflammation, and epithelial-mesenchymal transition. A meta-analysis combining our and public human transcriptomic datasets with murine models of NASH and fibrosis defined a molecular signature characterizing NASH and fibrosis and evidencing abnormal inflammation and extracellular matrix (ECM) homeostasis. Dermatopontin expression was found increased in fibrosis, and reversal of fibrosis after gastric bypass correlated with decreased dermatopontin expression. Functional studies in mice identified an active role for dermatopontin in collagen deposition and fibrosis. PPARα activation lowered dermatopontin expression through a transrepressive mechanism affecting the Klf6/TGFβ1 pathway. Liver fibrotic histological damages are thus characterized by the deregulated expression of a restricted set of inflammation- and ECM-related genes. Among them, dermatopontin may be a valuable target to reverse the hepatic fibrotic process.
Collapse
Affiliation(s)
- Philippe Lefebvre
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Fanny Lalloyer
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Eric Baugé
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Michal Pawlak
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Céline Gheeraert
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Hélène Dehondt
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Jonathan Vanhoutte
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Eloise Woitrain
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Nathalie Hennuyer
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Claire Mazuy
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Marie Bobowski-Gérard
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Francesco Paolo Zummo
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bruno Derudas
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | | | | | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, and.,Department of Endocrinology, Diabetology and Metabolism, University Hospital Antwerp, Edegem, Belgium
| | - Wilhelmus J Kwanten
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Peter Michielsen
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Thomas Vanwolleghem
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Jérôme Eeckhoute
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Antwerp, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Luc Van Gaal
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Antwerp, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Sven Francque
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Bart Staels
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| |
Collapse
|
27
|
Chang JZC, Hsieh YP, Lin WH, Chen HM, Kuo MYP. Activation of transforming growth factor-β1 by thrombin via integrins αvβ1, αvβ3, and αvβ5 in buccal fibroblasts: Suppression by epigallocatechin-3-gallate. Head Neck 2017; 39:1436-1445. [DOI: 10.1002/hed.24791] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 10/27/2016] [Accepted: 02/15/2017] [Indexed: 02/01/2023] Open
Affiliation(s)
- Jenny Zwei-Chieng Chang
- School of Dentistry; College of Medicine, National Taiwan University; Taipei Taiwan
- Department of Dentistry; National Taiwan University Hospital; Taipei Taiwan
| | - Yu-Ping Hsieh
- School of Dentistry; College of Medicine, National Taiwan University; Taipei Taiwan
| | - Wen-Hsin Lin
- School of Dentistry; College of Medicine, National Taiwan University; Taipei Taiwan
| | - Hsin-Ming Chen
- School of Dentistry; College of Medicine, National Taiwan University; Taipei Taiwan
- Department of Dentistry; National Taiwan University Hospital; Taipei Taiwan
| | - Mark Yen-Ping Kuo
- School of Dentistry; College of Medicine, National Taiwan University; Taipei Taiwan
- Department of Dentistry; National Taiwan University Hospital; Taipei Taiwan
| |
Collapse
|
28
|
Peng Y, Yan S, Chen D, Cui X, Jiao K. Pdgfrb is a direct regulatory target of TGFβ signaling in atrioventricular cushion mesenchymal cells. PLoS One 2017; 12:e0175791. [PMID: 28426709 PMCID: PMC5398542 DOI: 10.1371/journal.pone.0175791] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/31/2017] [Indexed: 12/30/2022] Open
Abstract
Cushion formation is the initial step for the development of valvuloseptal structures in mammalian hearts. TGFβ signaling plays critical roles in multiple steps of cushion morphogenesis. We used a newly developed conditional immortal atrioventricular cushion mesenchymal cell line, tsA58-AVM, to identify the TGFβ regulatory target genes through microarray analysis. Expression of ~1350 genes was significantly altered by TGFβ1 treatment. Subsequent bioinformatic analysis of TGFβ activated genes revealed that PDGF-BB signaling is the top hit as the potential upstream regulator. Among the 37 target molecules, 10 genes known to be involved in valve development and hemostasis were selected for quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis. Our results confirmed that they are all upregulated by TGFβ1 stimulation in tsA58-AVM cells and in primary atrioventricular cushion cells. We focused on examining regulation of Pdgfrb by TGFβ1, which encodes a tyrosine kinase receptor for PDGF-BB. We found that the ~150bp Pdgfrb promoter can respond to TGFβ stimulation and that this response relies on the two SP1 binding sites within the promoter. Co-immunoprecipitation analysis confirmed SP1 interacts with SMAD2 in a TGFβ-dependent fashion. Furthermore, SMAD2 is associated with the Pdgfrb promoter and this association is diminished by knocking down expression of Sp1. Our data therefore collectively suggest that upon TGFβ stimulation, SP1 recruits SMAD2 to the promoter of Pdgfrb to up-regulate its expression and thus Pdgfrb is a direct downstream target of the TGFβ/SMAD2 signaling.
Collapse
Affiliation(s)
- Yin Peng
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Shun Yan
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dongquan Chen
- Division of Preventive Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Xiangqin Cui
- Department of Biostatistics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Kai Jiao
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
29
|
Ebrahimi H, Naderian M, Sohrabpour AA. New Concepts on Pathogenesis and Diagnosis of Liver Fibrosis; A Review Article. Middle East J Dig Dis 2016; 8:166-178. [PMID: 27698966 PMCID: PMC5045669 DOI: 10.15171/mejdd.2016.29] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Liver fibrosis is a potentially reversible response to hepatic insults, triggered by different chronic diseases most importantly viral hepatitis, alcoholic, and nonalcoholic fatty liver disease. In the course of the chronic liver disease, hepatic fibrogenesis may develop, which is attributed to various types of cells, molecules, and pathways. Activated hepatic stellate cell (HSC), the primary source of extracellular matrix (ECM), is fundamental in pathophysiology of fibrogenesis, and thus is the most attractable target for reversing liver fibrosis. Although, liver biopsy has long been considered as the gold standard for diagnosis and staging of hepatic fibrosis, assessing progression and regression by biopsy is hampered by its limitations. We provide recent views on noninvasive approaches including serum biomarkers and radiologic techniques.
Collapse
Affiliation(s)
- Hedyeh Ebrahimi
- Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Naderian
- Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Sohrabpour
- Assistant Professor, Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Seo W, Jeong WI. Novel insight into a platelet-derived growth factor-C/Smad3 axis in liver fibrosis. Focus on "Role of Smad3 in platelet-derived growth factor-C-induced liver fibrosis". Am J Physiol Cell Physiol 2016; 310:C434-5. [PMID: 26791492 DOI: 10.1152/ajpcell.00369.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Wonhyo Seo
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| |
Collapse
|