1
|
Yaseen U, Hwang S, Park S, Kim SB, Lee HJ, Cha JY. New Insights into the Role of KLF10 in Tissue Fibrosis. Int J Mol Sci 2024; 25:1276. [PMID: 38279278 PMCID: PMC10816924 DOI: 10.3390/ijms25021276] [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: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Fibrosis, characterized by excessive extracellular matrix accumulation, disrupts normal tissue architecture, causes organ dysfunction, and contributes to numerous chronic diseases. This review focuses on Krüppel-like factor 10 (KLF10), a transcription factor significantly induced by transforming growth factor-β (TGF-β), and its role in fibrosis pathogenesis and progression across various tissues. KLF10, initially identified as TGF-β-inducible early gene-1 (TIEG1), is involved in key biological processes including cell proliferation, differentiation, apoptosis, and immune responses. Our analysis investigated KLF10 gene and protein structures, interaction partners, and context-dependent functions in fibrotic diseases. This review highlights recent findings that underscore KLF10 interaction with pivotal signaling pathways, such as TGF-β, and the modulation of gene expression in fibrotic tissues. We examined the dual role of KLF10 in promoting and inhibiting fibrosis depending on tissue type and fibrotic context. This review also discusses the therapeutic potential of targeting KLF10 in fibrotic diseases, based on its regulatory role in key pathogenic mechanisms. By consolidating current research, this review aims to enhance the understanding of the multifaceted role of KLF10 in fibrosis and stimulate further research into its potential as a therapeutic target in combating fibrotic diseases.
Collapse
Affiliation(s)
- Uzma Yaseen
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Soonjae Hwang
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| | - Sangbin Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Soo-Bin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Ho-Jae Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| | - Ji-Young Cha
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| |
Collapse
|
2
|
Thakkar C, Alikunju S, Niranjan N, Rizvi W, Abbas A, Abdellatif M, Sayed D. Klf9 plays a critical role in GR -dependent metabolic adaptations in cardiomyocytes. Cell Signal 2023; 111:110886. [PMID: 37690661 PMCID: PMC10591860 DOI: 10.1016/j.cellsig.2023.110886] [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: 06/02/2023] [Revised: 08/29/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Glucocorticoids through activation of the Glucocorticoid receptor (GR) play an essential role in cellular homeostasis during physiological variations and in response to stress. Our genomic GR binding and transcriptome data from Dexamethasone (Dex) treated cardiomyocytes showed an early differential regulation of mostly transcription factors, followed by sequential change in genes involved in downstream functional pathways. We examined the role of Krüppel-like factor 9 (Klf9), an early direct target of GR in cardiomyocytes. Klf9-ChIPseq identified 2150 genes that showed an increase in Klf9 binding in response to Dex. Transcriptome analysis of Dex treated cardiomyocytes with or without knockdown of Klf9 revealed differential regulation of 1777 genes, of which a reversal in expression is seen in 1640 genes with knockdown of Klf9 compared to Dex. Conversely, only 137 (∼8%) genes show further dysregulation in expression with siKLf9, as seen with Dex treated cardiomyocytes. Functional annotation identified genes of metabolic pathways on the top of differentially expressed genes, including those involved in glycolysis and oxidative phosphorylation. Knockdown of Klf9 in cardiomyocytes inhibited Dex induced increase in glycolytic function and mitochondrial spare respiratory capacity, as measured by glycolysis and mito stress tests, respectively. Thus, we conclude that cyclic, diurnal GR activation, through Klf9 -dependent feedforward signaling plays a central role in maintaining cellular homeostasis through metabolic adaptations in cardiomyocytes.
Collapse
Affiliation(s)
- Chandni Thakkar
- From the Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Saleena Alikunju
- From the Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Nandita Niranjan
- From the Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Wajiha Rizvi
- High School Research Intern, Wayne Hills High School, Wayne, NJ 07470, United States of America
| | - Ali Abbas
- From the Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ 07103, United States of America
| | - Maha Abdellatif
- From the Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Danish Sayed
- From the Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America.
| |
Collapse
|
3
|
Hwang S, Park S, Yaseen U, Lee HJ, Cha JY. KLF10 Inhibits TGF-β-Mediated Activation of Hepatic Stellate Cells via Suppression of ATF3 Expression. Int J Mol Sci 2023; 24:12602. [PMID: 37628783 PMCID: PMC10454374 DOI: 10.3390/ijms241612602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Liver fibrosis is a progressive and debilitating condition characterized by the excessive deposition of extracellular matrix proteins. Stellate cell activation, a major contributor to fibrogenesis, is influenced by Transforming growth factor (TGF-β)/SMAD signaling. Although Krüppel-like-factor (KLF) 10 is an early TGF-β-inducible gene, its specific role in hepatic stellate cell activation remains unclear. Our previous study demonstrated that KLF10 knockout mice develop severe liver fibrosis when fed a high-sucrose diet. Based on these findings, we aimed to identify potential target molecules involved in liver fibrosis and investigate the mechanisms underlying the KLF10 modulation of hepatic stellate cell activation. By RNA sequencing analysis of liver tissues from KLF10 knockout mice with severe liver fibrosis induced by a high-sucrose diet, we identified ATF3 as a potential target gene regulated by KLF10. In LX-2 cells, an immortalized human hepatic stellate cell line, KLF10 expression was induced early after TGF-β treatment, whereas ATF3 expression showed delayed induction. KLF10 knockdown in LX-2 cells enhanced TGF-β-mediated activation, as evidenced by elevated fibrogenic protein levels. Further mechanistic studies revealed that KLF10 knockdown promoted TGF-β signaling and upregulated ATF3 expression. Conversely, KLF10 overexpression suppressed TGF-β-mediated activation and downregulated ATF3 expression. Furthermore, treatment with the chemical chaperone 4-PBA attenuated siKLF10-mediated upregulation of ATF3 and fibrogenic responses in TGF-β-treated LX-2 cells. Collectively, our findings suggest that KLF10 acts as a negative regulator of the TGF-β signaling pathway, exerting suppressive effects on hepatic stellate cell activation and fibrogenesis through modulation of ATF3 expression. These results highlight the potential therapeutic implications of targeting the KLF10-ATF3 axis in liver fibrosis treatment.
Collapse
Affiliation(s)
- Soonjae Hwang
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (S.H.); (H.-J.L.)
| | - Sangbin Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (S.P.); (U.Y.)
| | - Uzma Yaseen
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (S.P.); (U.Y.)
| | - Ho-Jae Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (S.H.); (H.-J.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (S.P.); (U.Y.)
| | - Ji-Young Cha
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (S.H.); (H.-J.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (S.P.); (U.Y.)
| |
Collapse
|
4
|
Inhibition of Klf10 Attenuates Oxidative Stress-Induced Senescence of Chondrocytes via Modulating Mitophagy. Molecules 2023; 28:molecules28030924. [PMID: 36770589 PMCID: PMC9921806 DOI: 10.3390/molecules28030924] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease in the elderly. Accumulation of evidence has suggested that chondrocyte senescence plays a significant role in OA development. Here, we show that Krüppel-like factor 10 (Klf10), also named TGFβ inducible early gene-1 (TIEG1), is involved in the pathology of chondrocyte senescence. Knocking down the Klf10 in chondrocytes attenuated the tert-butyl hydroperoxide (TBHP)-induced senescence, inhibited generation of reactive oxygen species (ROS), and maintained mitochondrial homeostasis by activating mitophagy. These findings suggested that knocking down Klf10 inhibited senescence-related changes in chondrocytes and improved cartilage homeostasis, indicating that Klf10 may be a therapeutic target for protecting cartilage against OA.
Collapse
|
5
|
Yang EM, Park JS, Joo SY, Bae EH, Ma SK, Kim SW. Stanniocalcin‑1 suppresses TGF‑β‑induced mitochondrial dysfunction and cellular fibrosis in human renal proximal tubular cells. Int J Mol Med 2022; 50:107. [PMID: 35730604 DOI: 10.3892/ijmm.2022.5163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/29/2022] [Indexed: 12/30/2022] Open
Abstract
Stanniocalcin‑1 (STC1), a multifunctional glycoprotein with antioxidant and anti‑inflammatory properties, serves an important role in kidney protection. STC1 is one of the few hormones targeted to the mitochondria to regulate mitochondrial quality control by suppressing oxidative stress and mitochondrial damage. However, the mechanisms underlying the effect of STC1 remain unclear. The present study aimed to investigate the protective role of recombinant STC1 (rSTC1) in renal fibrosis and to identify the mechanisms underlying cellular fibrosis in HK2 human renal proximal tubular cells. Semi‑quantitative PCR, western blot analysis and confocal microscopy were used to detect the mRNA levels, protein levels and mitochondrial membrane potential (MMP). Mitochondrial superoxide production was evaluated using MitoSox staining. rSTC1 attenuated TGF‑β‑induced downregulation of AMP‑activated protein kinase and uncoupling protein 2 (UCP2). Treatment of HK2 cells with TGF‑β reduced the MMP and increased the production of reactive oxygen species (ROS). In addition, TGF‑β treatment upregulated fibrotic markers, such as α‑SMA and fibronectin, in HK2 cells. Treatment with rSTC1 and TGF‑β suppressed mitochondrial ROS production by recovering the MMP and reversed the upregulation of fibrotic markers in HK2 cells. The effects of rSTC1 were reversed when UCP2 expression was silenced. The present study revealed a novel role of STC1 in preventing TGF‑β induced cellular fibrosis in HK2 cells.
Collapse
Affiliation(s)
- Eun Mi Yang
- Department of Pediatrics, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jung Sun Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Soo Yeon Joo
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
6
|
Serum and Soleus Metabolomics Signature of Klf10 Knockout Mice to Identify Potential Biomarkers. Metabolites 2022; 12:metabo12060556. [PMID: 35736488 PMCID: PMC9231117 DOI: 10.3390/metabo12060556] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022] Open
Abstract
The transcription factor Krüppel-like factor 10 (Klf10), also known as Tieg1 for TGFβ (Inducible Early Gene-1) is known to control numerous genes in many cell types that are involved in various key biological processes (differentiation, proliferation, apoptosis, inflammation), including cell metabolism and human disease. In skeletal muscle, particularly in the soleus, deletion of the Klf10 gene (Klf10 KO) resulted in ultrastructure fiber disorganization and mitochondrial metabolism deficiencies, characterized by muscular hypertrophy. To determine the metabolic profile related to loss of Klf10 expression, we analyzed blood and soleus tissue using UHPLC-Mass Spectrometry. Metabolomics analyses on both serum and soleus revealed profound differences between wild-type (WT) and KO animals. Klf10 deficient mice exhibited alterations in metabolites associated with energetic metabolism. Additionally, chemical classes of aromatic and amino-acid compounds were disrupted, together with Krebs cycle intermediates, lipids and phospholipids. From variable importance in projection (VIP) analyses, the Warburg effect, citric acid cycle, gluconeogenesis and transfer of acetyl groups into mitochondria appeared to be possible pathways involved in the metabolic alterations observed in Klf10 KO mice. These studies have revealed essential roles for Klf10 in regulating multiple metabolic pathways whose alterations may underlie the observed skeletal muscle defects as well as other diseases.
Collapse
|
7
|
Kong HJ, Lee JJ, Kim JW, Kim J, Kim YO, Yeo SY. Zebrafish Klf11b is Required to Maintain Cell Viability by Inhibiting p53-Mediated Apoptosis. Dev Reprod 2022; 26:79-90. [PMID: 35950165 PMCID: PMC9336215 DOI: 10.12717/dr.2022.26.2.79] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/24/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
Krüppel-like factor 10 (KLF10) regulates various cellular functions, such as proliferation, differentiation and apoptosis, as well as the homeostasis of several types of tissue. In the present study, we attempted a loss-of-function analysis of zebrafish Klf11a and Klf11b, which constitute human KLF10 homologs. Embryos injected with klf11b-morpholino (MO) showed developmental retardation and cell death, whereas klf11a-MO-injected embryos showed normal development. In klf11b-MO-injected embryos, a dramatic increase in the amount of zebrafish p53 mRNA might be the cause of the increase in that of bax. The degree of apoptosis decreased in the klf11b-MO and p53-MO co-injected embryos. These findings imply that KLF10 is a negative regulator of p53-dependent transcription, suggesting that the KLF10/p53 complex may play an important role in apoptosis for maintenance of tissue homeostasis during embryonic development.
Collapse
Affiliation(s)
- Hee Jeong Kong
- Biotechnology Research Division, National Institute of Fisheries Science, Busan 46083, Korea
- Corresponding author Hee Jeong Kong, Biotechnology Research Division, National Institute of Fisheries Science, Busan 46083, Korea. Tel: +82-51-720-2455, Fax: +82-51-720-2456, E-mail: , Sang-Yeob Yeo, Department of Chemical and Biological Engineering, Hanbat National University, Daejeon 34158, Korea. Tel: +82-42-821-1552, Fax: +82-42-821-1692, E-mail:
| | - Jung Jin Lee
- Dept. of Chemical and Biological Engineering, Hanbat National University, Daejeon 34158, Korea
| | - Ju-Won Kim
- Biotechnology Research Division, National Institute of Fisheries Science, Busan 46083, Korea
| | - Julan Kim
- Biotechnology Research Division, National Institute of Fisheries Science, Busan 46083, Korea
| | - Young-Ok Kim
- Biotechnology Research Division, National Institute of Fisheries Science, Busan 46083, Korea
| | - Sang-Yeob Yeo
- Dept. of Chemical and Biological Engineering, Hanbat National University, Daejeon 34158, Korea
- Corresponding author Hee Jeong Kong, Biotechnology Research Division, National Institute of Fisheries Science, Busan 46083, Korea. Tel: +82-51-720-2455, Fax: +82-51-720-2456, E-mail: , Sang-Yeob Yeo, Department of Chemical and Biological Engineering, Hanbat National University, Daejeon 34158, Korea. Tel: +82-42-821-1552, Fax: +82-42-821-1692, E-mail:
| |
Collapse
|
8
|
Kammoun M, Nadal-Desbarats L, Même S, Lafoux A, Huchet C, Meyer-Dilhet G, Courchet J, Montigny F, Szeremeta F, Même W, Veksler V, Piquereau J, Pouletaut P, Subramaniam M, Hawse JR, Constans JM, Bensamoun SF. Deciphering the Role of Klf10 in the Cerebellum. JOURNAL OF BIOMEDICAL SCIENCE AND ENGINEERING 2022; 15:140-156. [PMID: 36507464 PMCID: PMC9733405 DOI: 10.4236/jbise.2022.155014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent studies have demonstrated a new role for Klf10, a Krüppel-like transcription factor, in skeletal muscle, specifically relating to mitochondrial function. Thus, it was of interest to analyze additional tissues that are highly reliant on optimal mitochondrial function such as the cerebellum and to decipher the role of Klf10 in the functional and structural properties of this brain region. In vivo (magnetic resonance imaging and localized spectroscopy, behavior analysis) and in vitro (histology, spectroscopy analysis, enzymatic activity) techniques were applied to comprehensively assess the cerebellum of wild type (WT) and Klf10 knockout (KO) mice. Histology analysis and assessment of locomotion revealed no significant difference in Klf10 KO mice. Diffusion and texture results obtained using MRI revealed structural changes in KO mice characterized as defects in the organization of axons. These modifications may be explained by differences in the levels of specific metabolites (myo-inositol, lactate) within the KO cerebellum. Loss of Klf10 expression also led to changes in mitochondrial activity as reflected by a significant increase in the activity of citrate synthase, complexes I and IV. In summary, this study has provided evidence that Klf10 plays an important role in energy production and mitochondrial function in the cerebellum.
Collapse
Affiliation(s)
- Malek Kammoun
- Biomechanics and Bioengineering CNRS UMR 7338, Sorbonne University—University of Technology of Compiègne, Compiègne, France
| | | | - Sandra Même
- Center for Molecular Biophysics, CNRS UPR4301, Orléans, France
| | - Aude Lafoux
- Therassay Platform, University of Nantes, Nantes, France
| | | | | | - Julien Courchet
- CNRS UMR-5310 and INSERM U-1217, NeuroMyoGène Institute, Villeurbanne, France
| | | | | | - William Même
- Center for Molecular Biophysics, CNRS UPR4301, Orléans, France
| | - Vladimir Veksler
- INSERM UMR-S 1180, University of Paris-Saclay, Châtenay-Malabry, France
| | - Jérôme Piquereau
- INSERM UMR-S 1180, University of Paris-Saclay, Châtenay-Malabry, France
| | - Philippe Pouletaut
- Biomechanics and Bioengineering CNRS UMR 7338, Sorbonne University—University of Technology of Compiègne, Compiègne, France
| | | | - John R. Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, USA
| | | | - Sabine F. Bensamoun
- Biomechanics and Bioengineering CNRS UMR 7338, Sorbonne University—University of Technology of Compiègne, Compiègne, France
| |
Collapse
|
9
|
Hsu YC, Ho C, Shih YH, Ni WC, Li YC, Chang HC, Lin CL. Knockout of KLF10 Ameliorated Diabetic Renal Fibrosis via Downregulation of DKK-1. Molecules 2022; 27:2644. [PMID: 35565995 PMCID: PMC9105565 DOI: 10.3390/molecules27092644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes-induced chronic kidney disease leads to mortality and morbidity and thus poses a great health burden worldwide. Krüppel-like factor 10 (KLF10), a zinc finger-containing transcription factor, regulates numerous cellular functions, such as proliferation, differentiation, and apoptosis. In this study, we explored the effects of KLF10 on diabetes-induced renal disease by using a KLF10 knockout mice model. Knockout of KLF10 obviously diminished diabetes-induced tumor growth factor-β (TGF-β), fibronectin, and type IV collagen expression, as evidenced by immunohistochemical staining. KLF10 knockout also repressed the expression of Dickkopf-1 (DKK-1) and phosphorylated β-catenin in diabetic mice, as evidenced by immunohistochemical staining and Western blot analysis. Quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) revealed that significantly decreased type IV collagen, fibronectin, and DKK-1 existed in KLF10 knockout diabetic mice compared with control diabetic mice. Moreover, knockout of KLF10 reduced the renal fibrosis, as shown by Masson's Trichrome analysis. Overall, the results indicate that depletion of KLF10 ameliorated diabetic renal fibrosis via the downregulation of DKK-1 expression and inhibited TGF-β1 and phosphorylated β-catenin expression. Our findings suggest that KLF10 may be a promising therapeutic choice for the treatment of diabetes-induced renal fibrosis.
Collapse
Affiliation(s)
- Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Cheng Ho
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Ya-Hsueh Shih
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Wen-Chiu Ni
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Yi-Chen Li
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Hsiu-Ching Chang
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
10
|
Memon A, Pyao Y, Jung Y, Choi HS, Song KD, Lee WK. The basal transcriptional activity of the murine Klf10 gene is regulated by the transcriptional factor JunB. Genes Genomics 2021; 43:343-349. [PMID: 33555508 DOI: 10.1007/s13258-020-01024-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Krüppel-like factor 10 (KLF10) belongs to the Sp1-like transcription factor family, which plays an important role in many directions, e.g., cell proliferation, apoptosis, and differentiation. Its 5' upstream regions are conserved across mammalian species. However, the regulatory mechanism has not been elucidated yet. OBJECTIVE Nonetheless the basal transcriptional regulation mechanisms of these regions are unknown. Here, we characterized it which is indispensable for the basal transcription of the Klf10 gene. METHODS Seven deletions of 5' upstream DNA fragments from the 10 kb mKlf10 genomic DNA were produced by PCR and cloned into the upstream of the luciferase (Luc) reporter gene in the pGL3 basic plasmid. RESULT The luciferase reporter assay showed that the DNA sequence at positions from -101 to +68 was required for a principle activity in the promoter of mKlf10 gene, in which transcriptional factor binding motifs, one JunB and two Sp1 sites, are included. Mutations at the sequence of JunB motif, but not at the two Sp1, abrogated the promoter activity completely, suggesting the indispensable role of JunB site for basal transcription of mKlf10 gene. Moreover, electrophoretic mobility and supershift assays (EMSA) uncovered that JunB protein bound to this region specifically. CONCLUSION Taken together, our study revealed that the JunB but not Sp1 at mKlf10 promoter functions as a positive basic factor for the transcriptional activity of the gene.
Collapse
Affiliation(s)
- Azra Memon
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon, 22212, South Korea
| | - Yuliya Pyao
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon, 22212, South Korea
| | - Yerin Jung
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon, 22212, South Korea
| | - Hwa-Sik Choi
- Department of Biomedical Laboratory Science, Shinhan University, Uijeongbu, 11644, South Korea
| | - Ki-Duk Song
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896, South Korea.
| | - Woon Kyu Lee
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon, 22212, South Korea.
| |
Collapse
|
11
|
Lee J, Oh AR, Lee HY, Moon YA, Lee HJ, Cha JY. Deletion of KLF10 Leads to Stress-Induced Liver Fibrosis upon High Sucrose Feeding. Int J Mol Sci 2020; 22:ijms22010331. [PMID: 33396939 PMCID: PMC7794950 DOI: 10.3390/ijms22010331] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a consequence of chronic liver injury associated with chronic viral infection, alcohol abuse, and nonalcoholic fatty liver. The evidence from clinical and animal studies indicates that transforming growth factor-β (TGF-β) signaling is associated with the development of liver fibrosis. Krüppel-like factor 10 (KLF10) is a transcription factor that plays a significant role in TGF-β-mediated cell growth, apoptosis, and differentiation. In recent studies, it has been reported to be associated with glucose homeostasis and insulin resistance. In the present study, we investigated the role of KLF10 in the progression of liver disease upon a high-sucrose diet (HSD) in mice. Wild type (WT) and Klf10 knockout (KO) mice were fed either a control chow diet or HSD (50% sucrose) for eight weeks. Klf10 KO mice exhibited significant hepatic steatosis, inflammation, and liver injury upon HSD feeding, whereas the WT mice exhibited mild hepatic steatosis with no apparent liver injury. The livers of HSD-fed Klf10 KO mice demonstrated significantly increased endoplasmic reticulum stress, oxidative stress, and proinflammatory cytokines. Klf10 deletion led to the development of sucrose-induced hepatocyte cell death both in vivo and in vitro. Moreover, it significantly increased fibrogenic gene expression and collagen accumulation in the liver. Increased liver fibrosis was accompanied by increased phosphorylation and nuclear localization of Smad3. Here, we demonstrate that HSD-fed mice develop a severe liver injury in the absence of KLF10 due to the hyperactivation of the endoplasmic reticulum stress response and CCAAT/enhance-binding protein homologous protein (CHOP)-mediated apoptosis of hepatocytes. The current study suggests that KLF10 plays a protective role against the progression of hepatic steatosis into liver fibrosis in a lipogenic state.
Collapse
Affiliation(s)
- Junghoon Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Ah-Reum Oh
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Hui-Young Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University School of Medicine, Incheon 22212, Korea;
| | - Ho-Jae Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
- Correspondence: (H.-J.L.); (J.-Y.C.); Tel.: +82-32-899-6054 (H.-J.L.); +82-32-899-6070 (J.-Y.C.)
| | - Ji-Young Cha
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
- Gachon Medical Research Institute, Gachon University Gil Medical Center, Incheon 21565, Korea
- Correspondence: (H.-J.L.); (J.-Y.C.); Tel.: +82-32-899-6054 (H.-J.L.); +82-32-899-6070 (J.-Y.C.)
| |
Collapse
|
12
|
Gurung S, Ulrich D, Sturm M, Rosamilia A, Werkmeister JA, Gargett CE. Comparing the Effect of TGF-β Receptor Inhibition on Human Perivascular Mesenchymal Stromal Cells Derived from Endometrium, Bone Marrow and Adipose Tissues. J Pers Med 2020; 10:jpm10040261. [PMID: 33271899 PMCID: PMC7712261 DOI: 10.3390/jpm10040261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Rare perivascular mesenchymal stromal cells (MSCs) with therapeutic properties have been identified in many tissues. Their rarity necessitates extensive in vitro expansion, resulting in spontaneous differentiation, cellular senescence and apoptosis, producing therapeutic products with variable quality and decreased potency. We previously demonstrated that A83-01, a transforming growth factor beta (TGF-β) receptor inhibitor, maintained clonogenicity and promoted the potency of culture-expanded premenopausal endometrial MSCs using functional assays and whole-transcriptome sequencing. Here, we compared the effects of A83-01 on MSCs derived from postmenopausal endometrium, menstrual blood, placenta decidua-basalis, bone marrow and adipose tissue. Sushi-domain-containing-2 (SUSD2+) and CD34+CD31−CD45− MSCs were isolated. Expanded MSCs were cultured with or without A83-01 for 7 days and assessed for MSC properties. SUSD2 identified perivascular cells in the placental decidua-basalis, and their maternal origin was validated. A83-01 promoted MSC proliferation from all sources except bone marrow and only increased SUSD2 expression and prevented apoptosis in MSCs from endometrial-derived tissues. A83-01 only improved the cloning efficiency of postmenopausal endometrial MSCs (eMSCs), and expanded adipose tissue MSCs (adMSCs) underwent significant senescence, which was mitigated by A83-01. MSCs derived from bone marrow (bmMSCs) were highly apoptotic, but A83-01 was without effect. A83-01 maintained the function and phenotype in MSCs cultured from endometrial, but not other, tissues. Our results also demonstrated that cellular SUSD2 expression directly correlates with the functional phenotype.
Collapse
Affiliation(s)
- Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
- Correspondence: ; Tel.: +61-03-8572-2813
| | - Daniela Ulrich
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz, 8036 Graz, Austria
| | - Marian Sturm
- Cell & Tissue Therapies WA, Royal Perth Hospital, Perth, WA 6000, Australia;
- Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Perth, WA 6009, Australia
| | - Anna Rosamilia
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
- Monash Health, Clayton, VIC 3168, Australia
| | - Jerome A. Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
| |
Collapse
|
13
|
Mal R, Magner A, David J, Datta J, Vallabhaneni M, Kassem M, Manouchehri J, Willingham N, Stover D, Vandeusen J, Sardesai S, Williams N, Wesolowski R, Lustberg M, Ganju RK, Ramaswamy B, Cherian MA. Estrogen Receptor Beta (ERβ): A Ligand Activated Tumor Suppressor. Front Oncol 2020; 10:587386. [PMID: 33194742 PMCID: PMC7645238 DOI: 10.3389/fonc.2020.587386] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) belong to a superfamily of nuclear receptors called steroid hormone receptors, which, upon binding ligand, dimerize and translocate to the nucleus where they activate or repress the transcription of a large number of genes, thus modulating critical physiologic processes. ERβ has multiple isoforms that show differing association with prognosis. Expression levels of the full length ERβ1 isoform are often lower in aggressive cancers as compared to normal tissue. High ERβ1 expression is associated with improved overall survival in women with breast cancer. The promise of ERβ activation, as a potential targeted therapy, is based on concurrent activation of multiple tumor suppressor pathways with few side effects compared to chemotherapy. Thus, ERβ is a nuclear receptor with broad-spectrum tumor suppressor activity, which could serve as a potential treatment target in a variety of human cancers including breast cancer. Further development of highly selective agonists that lack ERα agonist activity, will be necessary to fully harness the potential of ERβ.
Collapse
Affiliation(s)
- Rahul Mal
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Alexa Magner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Joel David
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Jharna Datta
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Meghna Vallabhaneni
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Mahmoud Kassem
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jasmine Manouchehri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Natalie Willingham
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Daniel Stover
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jeffery Vandeusen
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Sagar Sardesai
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Nicole Williams
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Robert Wesolowski
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Maryam Lustberg
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Ramesh K Ganju
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Mathew A Cherian
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Leclère PS, Rousseau D, Patouraux S, Guérin S, Bonnafous S, Gréchez-Cassiau A, Ruberto AA, Luci C, Subramaniam M, Tran A, Delaunay F, Gual P, Teboul M. MCD diet-induced steatohepatitis generates a diurnal rhythm of associated biomarkers and worsens liver injury in Klf10 deficient mice. Sci Rep 2020; 10:12139. [PMID: 32699233 PMCID: PMC7376252 DOI: 10.1038/s41598-020-69085-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
A large number of hepatic functions are regulated by the circadian clock and recent evidence suggests that clock disruption could be a risk factor for liver complications. The circadian transcription factor Krüppel like factor 10 (KLF10) has been involved in liver metabolism as well as cellular inflammatory and death pathways. Here, we show that hepatic steatosis and inflammation display diurnal rhythmicity in mice developing steatohepatitis upon feeding with a methionine and choline deficient diet (MCDD). Core clock gene mRNA oscillations remained mostly unaffected but rhythmic Klf10 expression was abolished in this model. We further show that Klf10 deficient mice display enhanced liver injury and fibrosis priming upon MCDD challenge. Silencing Klf10 also sensitized primary hepatocytes to apoptosis along with increased caspase 3 activation in response to TNFα. This data suggests that MCDD induced steatohepatitis barely affects the core clock mechanism but leads to a reprogramming of circadian gene expression in the liver in analogy to what is observed in other experimental disease paradigms. We further identify KLF10 as a component of this transcriptional reprogramming and a novel hepato-protective factor.
Collapse
Affiliation(s)
- Pierre S Leclère
- Université Côte D'Azur, CNRS, INSERM, iBV, Nice, France.,Université Côte D'Azur, INSERM, C3M, Nice, France
| | | | | | - Sophie Guérin
- Université Côte D'Azur, CNRS, INSERM, iBV, Nice, France
| | | | | | | | - Carmelo Luci
- Université Côte D'Azur, INSERM, C3M, Nice, France
| | | | - Albert Tran
- Université Côte D'Azur, CHU, INSERM, C3M, Nice, France
| | | | | | - Michèle Teboul
- Université Côte D'Azur, CNRS, INSERM, iBV, Nice, France.
| |
Collapse
|
15
|
Latifi Z, Nejabati HR, Abroon S, Mihanfar A, Farzadi L, Hakimi P, Hajipour H, Nouri M, Fattahi A. Dual role of TGF-β in early pregnancy: clues from tumor progression. Biol Reprod 2020; 100:1417-1430. [PMID: 30772900 DOI: 10.1093/biolre/ioz024] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/25/2018] [Accepted: 02/14/2019] [Indexed: 01/10/2023] Open
Abstract
TGF-β signaling in the endometrium is active during the implantation period and has a pivotal role in regulating endometrial receptivity and embryo implantation. During embryo implantation, both apoptosis and proliferation of endometrial cells happen at the same time and it seems TGF-β is the factor that controls both of these processes. As shown in cancer cells, in special conditions this cytokine can have a dual effect and switch the action from apoptosis to proliferation. Owing to the similarity between embryo implantation and cancer development and also unusual pattern of proliferation and remodeling in the uterus, in this review we suggest the existence of such a switching in endometrium during the early pregnancy. Moreover, we address some potential mechanisms that could regulate the switching. A better understanding of the molecular mechanisms regulating TGF-β action and signaling during the implantation period could pave the way for introducing novel therapeutic strategies in order to solve implantation-associated issues such as repeated implantation failure.
Collapse
Affiliation(s)
- Zeinab Latifi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Abroon
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aynaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Laya Farzadi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Hakimi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hajipour
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Lin CL, Hsu YC, Huang YT, Shih YH, Wang CJ, Chiang WC, Chang PJ. A KDM6A-KLF10 reinforcing feedback mechanism aggravates diabetic podocyte dysfunction. EMBO Mol Med 2020; 11:emmm.201809828. [PMID: 30948420 PMCID: PMC6505577 DOI: 10.15252/emmm.201809828] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Diabetic nephropathy is the leading cause of end‐stage renal disease. Although dysfunction of podocytes, also termed glomerular visceral epithelial cells, is critically associated with diabetic nephropathy, the mechanism underlying podocyte dysfunction still remains obscure. Here, we identify that KDM6A, a histone lysine demethylase, reinforces diabetic podocyte dysfunction by creating a positive feedback loop through up‐regulation of its downstream target KLF10. Overexpression of KLF10 in podocytes not only represses multiple podocyte‐specific markers including nephrin, but also conversely increases KDM6A expression. We further show that KLF10 inhibits nephrin expression by directly binding to the gene promoter together with the recruitment of methyltransferase Dnmt1. Importantly, inactivation or knockout of either KDM6A or KLF10 in mice significantly suppresses diabetes‐induced proteinuria and kidney injury. Consistent with the notion, we also show that levels of both KDM6A and KLF10 proteins or mRNAs are substantially elevated in kidney tissues or in urinary exosomes of human diabetic nephropathy patients as compared with control subjects. Our findings therefore suggest that targeting the KDM6A–KLF10 feedback loop may be beneficial to attenuate diabetes‐induced kidney injury.
Collapse
Affiliation(s)
- Chun-Liang Lin
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Kidney Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi, Taiwan.,Center for Shockwave Medicine and Tissue Engineering, Department of Medical Research, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yung-Chien Hsu
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Ting Huang
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ya-Hsueh Shih
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ching-Jen Wang
- Center for Shockwave Medicine and Tissue Engineering, Department of Medical Research, Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wen-Chih Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Pey-Jium Chang
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan .,Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
17
|
Skugor A, Kjos NP, Sundaram AYM, Mydland LT, Ånestad R, Tauson AH, Øverland M. Effects of long-term feeding of rapeseed meal on skeletal muscle transcriptome, production efficiency and meat quality traits in Norwegian Landrace growing-finishing pigs. PLoS One 2019; 14:e0220441. [PMID: 31390356 PMCID: PMC6685631 DOI: 10.1371/journal.pone.0220441] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/16/2019] [Indexed: 12/30/2022] Open
Abstract
This study was performed to investigate the effects of dietary inclusion of 20% rapeseed meal (RSM) as an alternative to soybean meal (SBM) in a three-month feeding experiment with growing finishing pigs. Dietary alteration affected growth performance, several carcass traits and transcriptional responses in the skeletal muscle, but did not affect measured meat quality traits. In general, pigs fed the RSM test diet exhibited reduced growth performance compared to pigs on SBM control diet. Significant transcriptional changes in the skeletal muscle of growing pigs fed RSM diet were likely the consequence of an increased amount of fiber and higher polyunsaturated fatty acids, and presence of bioactive phytochemicals, such as glucosinolates. RNAseq pipeline using Tophat2-Cuffdiff identified 57 upregulated and 63 downregulated genes in RSM compared to SBM pigs. Significantly enriched among downregulated pathways was p53-mediated signalling involved in cellular proliferation, while activation of negative growth regulators (IER5, KLF10, BTG2, KLF11, RETREG1, PRUNE2) in RSM fed pigs provided further evidence for reduced proliferation and increased cellular death, in accordance with the observed reduction in performance traits. Upregulation of well-known metabolic controllers (PDK4, UCP3, ESRRG and ESRRB), involved in energy homeostasis (glucose and lipid metabolism, and mitochondrial function), suggested less available energy and nutrients in RSM pigs. Furthermore, several genes supported more pronounced proteolysis (ABTB1, OTUD1, PADI2, SPP1) and reduced protein synthesis (THBS1, HSF4, AP1S2) in RSM muscle tissue. In parallel, higher levels of NR4A3, PDK4 and FGF21, and a drop in adropin, ELOVL6 and CIDEC/FSP27 indicated increased lipolysis and fatty acid oxidation, reflective of lower dressing percentage. Finally, pigs exposed to RSM showed greater expression level of genes responsive to oxidative stress, indicated by upregulation of GPX1, GPX2, and TXNIP.
Collapse
Affiliation(s)
- Adrijana Skugor
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| | - Nils Petter Kjos
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| | | | - Liv Torunn Mydland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| | - Ragnhild Ånestad
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| | - Anne-Helene Tauson
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Margareth Øverland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| |
Collapse
|
18
|
Amerizadeh F, Bahrami A, Khazaei M, Hesari A, Rezayi M, Talebian S, Maftouh M, Moetamani-Ahmadi M, Seifi S, Shahidsales S, Joudi-Mashhad M, Ferns GA, Ghasemi F, Avan A. Current status and future prospects of transforming growth factor-β as a potential prognostic and therapeutic target in the treatment of breast cancer. J Cell Biochem 2019; 120:6962-6971. [PMID: 30672016 DOI: 10.1002/jcb.27831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/14/2018] [Indexed: 01/24/2023]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway is one of the important pathways involved in the cancer cell proliferation, invasion, migration, angiogenesis, apoptosis, as well as in metastasis by agitation or invasion of metastasis-related factors, including matrix metalloproteinase (MMP), epithelial-to-mesenchymal transition (EMT), tumor microenvironment (TME), cancer stem cells (CSCs), and cell adhesion molecules (CAMs). These data suggest its potential value as a therapeutic object in the treatment of malignancies including breast cancer. Several pharmacological approaches have been established to suppress TGF-β pathway; such as vaccines, small molecular inhibitors, antisense oligonucleotides, and monoclonal antibodies. Some of these are now approved by the US Food and Drug Administration for targeting the TGF-β signaling pathway. This study attempts to summarize the current data about the functions of TGF-β in cancer cells, and their probable application in the cancer therapy with a specific emphasis on recent preclinical and clinical research in the treatment of breast cancer and its prognostic value.
Collapse
Affiliation(s)
- Forouzan Amerizadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - AmirReza Hesari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezayi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Talebian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sima Seifi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mona Joudi-Mashhad
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, UK
| | - Faezeh Ghasemi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Single Nucleotide Polymorphism in SMAD7 and CHI3L1 and Colorectal Cancer Risk. Mediators Inflamm 2018; 2018:9853192. [PMID: 30498395 PMCID: PMC6222239 DOI: 10.1155/2018/9853192] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/01/2018] [Accepted: 08/16/2018] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the leading cancers throughout the world. It represents the third most common cancer and the fourth in mortality. Most of CRC are sporadic, arise with no known high-penetrant genetic variation and with no previous family history. The etiology of sporadic CRC is considered to be multifactorial and arises from the interaction of genetic variants of low-penetrant genes and environmental risk factors. The most common well-studied genetic variation is single nucleotide polymorphisms (SNPs). SNP arises as a point mutation. If the frequency of the sequence variation reaches 1% or more in the population, it is referred to as polymorphism, but if it is lower than 1%, the allele is typically considered as a mutation. Lots of SNPs have been associated with CRC development and progression, for example, genes of TGF-β1 and CHI3L1 pathways. TGF-β1 is a pleiotropic cytokine with a dual role in cancer development and progression. TGF-β1 mediates its actions through canonical and noncanonical pathways. The most important negative regulatory protein for TGF-β1 activity is termed SMAD7. The production of TGF-β can be controlled by another protein called YKL-40. YKL-40 is a glycoprotein with an important role in cancer initiation and metastasis. YKL-40 is encoded by the CHI3L1 gene. The aim of the present review is to give a brief introduction of CRC, SNP, and examples of some SNPs that have been documented to be associated with CRC. We also discuss two important signaling pathways TGF-β1 and CHI3L1 that influence the incidence and progression of CRC.
Collapse
|
20
|
Unverricht-Yeboah M, Giesen U, Kriehuber R. Comparative gene expression analysis after exposure to 123I-iododeoxyuridine, γ- and α-radiation-potential biomarkers for the discrimination of radiation qualities. JOURNAL OF RADIATION RESEARCH 2018; 59:411-429. [PMID: 29800458 PMCID: PMC6054186 DOI: 10.1093/jrr/rry038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/29/2017] [Indexed: 05/27/2023]
Abstract
Gene expression analysis was carried out in Jurkat cells in order to identify candidate genes showing significant gene expression alterations allowing robust discrimination of the Auger emitter 123I, incorporated into the DNA as 123I-iododeoxyuridine (123IUdR), from α- and γ-radiation. The γ-H2AX foci assay was used to determine equi-effect doses or activity, and gene expression analysis was carried out at similar levels of foci induction. Comparative gene expression analysis was performed employing whole human genome DNA microarrays. Candidate genes had to show significant expression changes and no altered gene regulation or opposite regulation after exposure to the radiation quality to be compared. The gene expression of all candidate genes was validated by quantitative real-time PCR. The functional categorization of significantly deregulated genes revealed that chromatin organization and apoptosis were generally affected. After exposure to 123IUdR, α-particles and γ-rays, at equi-effect doses/activity, 155, 316 and 982 genes were exclusively regulated, respectively. Applying the stringent requirements for candidate genes, four (PPP1R14C, TNFAIP8L1, DNAJC1 and PRTFDC1), one (KLF10) and one (TNFAIP8L1) gene(s) were identified, respectively allowing reliable discrimination between γ- and 123IUdR exposure, γ- and α-radiation, and α- and 123IUdR exposure, respectively. The Auger emitter 123I induced specific gene expression patterns in Jurkat cells when compared with γ- and α-irradiation, suggesting a unique cellular response after 123IUdR exposure. Gene expression analysis might be an effective tool for identifying biomarkers for discriminating different radiation qualities and, furthermore, might help to explain the varying biological effectiveness at the mechanistic level.
Collapse
Affiliation(s)
- Marcus Unverricht-Yeboah
- Radiation Biology Unit, Department of Safety and Radiation Protection, Forschungszentrum Jülich, Jülich, Germany
| | - Ulrich Giesen
- Physikalisch-Technische Bundesanstalt (PTB), Bundesallee 100, Braunschweig, Germany
| | - Ralf Kriehuber
- Radiation Biology Unit, Department of Safety and Radiation Protection, Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
21
|
Memon A, Lee WK. KLF10 as a Tumor Suppressor Gene and Its TGF-β Signaling. Cancers (Basel) 2018; 10:E161. [PMID: 29799499 PMCID: PMC6025274 DOI: 10.3390/cancers10060161] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/15/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022] Open
Abstract
Krüppel-like factor 10 (KLF10), originally named TGF-β (Transforming growth factor beta) inducible early gene 1 (TIEG1), is a DNA-binding transcriptional regulator containing a triple C2H2 zinc finger domain. By binding to Sp1 (specificity protein 1) sites on the DNA and interactions with other regulatory transcription factors, KLF10 encourages and suppresses the expression of multiple genes in many cell types. Many studies have investigated its signaling cascade, but other than the TGF-β/Smad signaling pathway, these are still not clear. KLF10 plays a role in proliferation, differentiation as well as apoptosis, just like other members of the SP (specificity proteins)/KLF (Krüppel-like Factors). Recently, several studies reported that KLF10 KO (Knock out) is associated with defects in cell and organs such as osteopenia, abnormal tendon or cardiac hypertrophy. Since KLF10 was first discovered, several studies have defined its role in cancer as a tumor suppressor. KLF10 demonstrate anti-proliferative effects and induce apoptosis in various carcinoma cells including pancreatic cancer, leukemia, and osteoporosis. Collectively, these data indicate that KLF10 plays a significant role in various biological processes and diseases, but its role in cancer is still unclear. Therefore, this review was conducted to describe and discuss the role and function of KLF10 in diseases, including cancer, with a special emphasis on its signaling with TGF-β.
Collapse
Affiliation(s)
- Azra Memon
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea.
| | - Woon Kyu Lee
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea.
| |
Collapse
|
22
|
Subramaniam M, Pitel KS, Bruinsma ES, Monroe DG, Hawse JR. TIEG and estrogen modulate SOST expression in the murine skeleton. J Cell Physiol 2017; 233:3540-3551. [PMID: 29044507 DOI: 10.1002/jcp.26211] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 09/26/2017] [Indexed: 01/02/2023]
Abstract
TIEG knockout (KO) mice exhibit a female-specific osteopenic phenotype and altered expression of TIEG in humans is associated with osteoporosis. Gene expression profiling studies identified sclerostin as one of the most highly up-regulated transcripts in the long bones of TIEG KO mice relative to WT littermates suggesting that TIEG may regulate SOST expression. TIEG was shown to substantially suppress SOST promoter activity and the regulatory elements through which TIEG functions were identified using promoter deletion and chromatin immunoprecipitation assays. Knockdown of TIEG in IDG-SW3 osteocyte cells using shRNA and CRISPR-Cas9 technology resulted in increased SOST expression and delayed mineralization, mimicking the results obtained from TIEG KO mouse bones. Given that TIEG is an estrogen regulated gene, and as changes in the hormonal milieu affect SOST expression, we performed ovariectomy (OVX) and estrogen replacement therapy (ERT) studies in WT and TIEG KO mice followed by miRNA and mRNA sequencing of cortical and trabecular compartments of femurs. SOST expression levels were considerably higher in cortical bone compared to trabecular bone. In cortical bone, SOST expression was increased following OVX only in WT mice and was suppressed following ERT in both genotypes. In contrast, SOST expression in trabecular bone was decreased following OVX and significantly increased following ERT. Interestingly, a number of miRNAs that are predicted to target sclerostin exhibited inverse expression levels in response to OVX and ERT. These data implicate important roles for TIEG and estrogen-regulated miRNAs in modulating SOST expression in bone.
Collapse
Affiliation(s)
| | - Kevin S Pitel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Elizabeth S Bruinsma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - David G Monroe
- Robert and Arlene Kogod Center on Aging and Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
23
|
Llontop P, Lopez-Fernandez D, Clavo B, Afonso Martín JL, Fiuza-Pérez MD, García Arranz M, Calatayud J, Molins López-Rodó L, Alshehri K, Ayub A, Raad W, Bhora F, Santana-Rodríguez N. Airway transplantation of adipose stem cells protects against bleomycin-induced pulmonary fibrosis. J Investig Med 2017; 66:739-746. [PMID: 29167193 DOI: 10.1136/jim-2017-000494] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2017] [Indexed: 12/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease with poor prognosis. Adipose-derived stem cells (ADSC) have demonstrated regenerative properties in several tissues. The hypothesis of this study was that airway transplantation of ADSC could protect against bleomycin (BLM)-induced pulmonary fibrosis (PF). Fifty-eight lungs from 29 male Sprague-Dawley rats were analyzed. Animals were randomly divided into five groups: a) control (n=3); b) sham (n=6); c) BLM (n=6); d) BLM+ADSC-2d (n=6); and e) BLM+ADSC-14d (n=8). Animals received 500 µL saline (sham), 2.5 UI/kg BLM in 500 µL saline (BLM), and 2×106 ADSC in 100 µL saline intratracheally at 2 (BLM+ADSC-2d) and 14 days (BLM+ADSC-14d) after BLM. Animals were sacrificed at 28 days. Blinded Ashcroft score was used to determine pulmonary fibrosis extent on histology. Hsp27, Vegf, Nfkβ, IL-1, IL-6, Col4, and Tgfβ1 mRNA gene expression were determined using real-time quantitative-PCR. Ashcroft index was: control=0; sham=0.37±0.07; BLM=6.55±0.34 vs sham (P=0.006). BLM vs BLM+ADSC-2d=4.63±0.38 (P=0.005) and BLM+ADSC-14d=3.77±0.46 (P=0.005). BLM vs sham significantly increased Hsp27 (P=0.018), Nfkβ (P=0.009), Col4 (P=0.004), Tgfβ1 (P=0.006) and decreased IL-1 (P=0.006). BLM+ADSC-2d vs BLM significantly decreased Hsp27 (P=0.009) and increased Vegf (P=0.006), Nfkβ (P=0.009). BLM+ADSC-14d vs BLM significantly decreased Hsp27 (P=0.028), IL-6 (P=0.013), Col4 (P=0.002), and increased Nfkβ (P=0.040) and Tgfβ1 (P=0.002). Airway transplantation of ADSC significantly decreased the fibrosis rate in both early and established pulmonary fibrosis, modulating the expression of Hsp27, Vegfa, Nfkβ, IL-6, Col4, and Tgfβ1. From a translational perspective, this technique could become a new adjuvant treatment for patients with IPF.
Collapse
Affiliation(s)
- Pedro Llontop
- Facultad de Psicología, Universidad Nacional de Educación a Distancia, Madrid, Spain.,Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS)-BioPharm Group, Universidadde Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr Negrín, Las Palmas de Gran Canaria, Spain.,Experimental Surgery and Medicine Unit, Hospital General Gregorio Marañon. Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain
| | - Daniel Lopez-Fernandez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS)-BioPharm Group, Universidadde Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr Negrín, Las Palmas de Gran Canaria, Spain
| | - Bernardino Clavo
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS)-BioPharm Group, Universidadde Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr Negrín, Las Palmas de Gran Canaria, Spain
| | - Juan Luis Afonso Martín
- Pathology Service, Complejo Hospitalario Materno Infantil, Las Palmas de Gran Canaria, Spain
| | - María D Fiuza-Pérez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS)-BioPharm Group, Universidadde Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr Negrín, Las Palmas de Gran Canaria, Spain
| | - Mariano García Arranz
- Department of Surgery, Laboratorio de Nuevas Tecnologías, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Joaquín Calatayud
- Department of Thoracic Surgery, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Khalid Alshehri
- Department of Thoracic Surgery, Mount Sinai Health System, New York, USA
| | - Adil Ayub
- Department of Thoracic Surgery, Mount Sinai Health System, New York, USA
| | - Wissam Raad
- Department of Thoracic Surgery, Mount Sinai Health System, New York, USA
| | - Faiz Bhora
- Department of Thoracic Surgery, Mount Sinai Health System, New York, USA
| | - Norberto Santana-Rodríguez
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS)-BioPharm Group, Universidadde Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Department of Thoracic Surgery, Mount Sinai Health System, New York, USA.,Section of Thoracic Surgery, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Dichotomous roles of TGF-β in human cancer. Biochem Soc Trans 2017; 44:1441-1454. [PMID: 27911726 DOI: 10.1042/bst20160065] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-β (TGF-β) mediates numerous biological processes, including embryonic development and the maintenance of cellular homeostasis in a context-dependent manner. Consistent with its central role in maintaining cellular homeostasis, inhibition of TGF-β signaling results in disruption of normal homeostatic processes and subsequent carcinogenesis, defining the TGF-β signaling pathway as a tumor suppressor. However, once carcinogenesis is initiated, the TGF-β signaling pathway promotes cancer progression. This dichotomous function of the TGF-β signaling pathway is mediated through altering effects on both the cancer cells, by inducing apoptosis and inhibiting proliferation, and the tumor microenvironment, by promoting angiogenesis and inhibiting immunosurveillance. Current studies support inhibition of TGF-β signaling either alone, or in conjunction with anti-angiogenic therapy or immunotherapy as a promising strategy for the treatment of human cancers.
Collapse
|
25
|
Kim CK, He P, Bialkowska AB, Yang VW. SP and KLF Transcription Factors in Digestive Physiology and Diseases. Gastroenterology 2017; 152:1845-1875. [PMID: 28366734 PMCID: PMC5815166 DOI: 10.1053/j.gastro.2017.03.035] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/14/2022]
Abstract
Specificity proteins (SPs) and Krüppel-like factors (KLFs) belong to the family of transcription factors that contain conserved zinc finger domains involved in binding to target DNA sequences. Many of these proteins are expressed in different tissues and have distinct tissue-specific activities and functions. Studies have shown that SPs and KLFs regulate not only physiological processes such as growth, development, differentiation, proliferation, and embryogenesis, but pathogenesis of many diseases, including cancer and inflammatory disorders. Consistently, these proteins have been shown to regulate normal functions and pathobiology in the digestive system. We review recent findings on the tissue- and organ-specific functions of SPs and KLFs in the digestive system including the oral cavity, esophagus, stomach, small and large intestines, pancreas, and liver. We provide a list of agents under development to target these proteins.
Collapse
Affiliation(s)
- Chang-Kyung Kim
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY
| | - Ping He
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY
| | - Agnieszka B. Bialkowska
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY,Corresponding Authors: Vincent W. Yang & Agnieszka B. Bialkowska, Department of Medicine, Stony Brook University School of Medicine, HSC T-16, Rm. 020; Stony Brook, NY, USA. Tel: (631) 444-2066; Fax: (631) 444-3144; ;
| | - Vincent W. Yang
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY,Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY,Corresponding Authors: Vincent W. Yang & Agnieszka B. Bialkowska, Department of Medicine, Stony Brook University School of Medicine, HSC T-16, Rm. 020; Stony Brook, NY, USA. Tel: (631) 444-2066; Fax: (631) 444-3144; ;
| |
Collapse
|
26
|
Kammoun M, Meme S, Meme W, Subramaniam M, Hawse JR, Canon F, Bensamoun SF. Impact of TIEG1 on the structural properties of fast- and slow-twitch skeletal muscle. Muscle Nerve 2016; 55:410-416. [PMID: 27421714 DOI: 10.1002/mus.25252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2016] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Transforming growth factor-beta (TGF-β)-inducible early gene-1 (TIEG1) is a transcription factor that is highly expressed in skeletal muscle. The purpose of this study was to characterize the structural properties of both fast-twitch (EDL) and slow-twitch (soleus) muscles in the hindlimb of TIEG1-deficient (TIEG1-/- ) mice. METHODS Ten slow and 10 fast muscles were analyzed from TIEG1-/- and wild-type (WT) mice using MRI texture (MRI-TA) and histological analyses. RESULTS MRI-TA could discriminate between WT slow and fast muscles. Deletion of the TIEG1 gene led to changes in the texture profile within both muscle types. Specifically, muscle isolated from TIEG1-/- mice displayed hypertrophy, hyperplasia, and a modification of fiber area distribution. CONCLUSIONS We demonstrated that TIEG1 plays an important role in the structural properties of skeletal muscle. This study further implicates important roles for TIEG1 in the development of skeletal muscle and suggests that defects in TIEG1 expression and/or function may be associated with muscle disease. Muscle Nerve 55: 410-416, 2017.
Collapse
Affiliation(s)
- Malek Kammoun
- Université de Technologie de Compiègne, Centre de Recherches de Royallieu, Laboratoire de Biomécanique et de BioIngénierie, UMR CNRS 7338, BP 20529, 60205, Compiègne Cedex, France
| | - Sandra Meme
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - William Meme
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - Malayannan Subramaniam
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Francis Canon
- Université de Technologie de Compiègne, Centre de Recherches de Royallieu, Laboratoire de Biomécanique et de BioIngénierie, UMR CNRS 7338, BP 20529, 60205, Compiègne Cedex, France
| | - Sabine F Bensamoun
- Université de Technologie de Compiègne, Centre de Recherches de Royallieu, Laboratoire de Biomécanique et de BioIngénierie, UMR CNRS 7338, BP 20529, 60205, Compiègne Cedex, France
| |
Collapse
|
27
|
Kammoun M, Pouletaut P, Canon F, Subramaniam M, Hawse JR, Vayssade M, Bensamoun SF. Impact of TIEG1 Deletion on the Passive Mechanical Properties of Fast and Slow Twitch Skeletal Muscles in Female Mice. PLoS One 2016; 11:e0164566. [PMID: 27736981 PMCID: PMC5063386 DOI: 10.1371/journal.pone.0164566] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/27/2016] [Indexed: 11/24/2022] Open
Abstract
As transforming growth factor (TGF)-β inducible early gene-1 is highly expressed in skeletal muscle, the effect of TIEG1 gene deletion on the passive mechanical properties of slow and fast twitch muscle fibers was analyzed. Twenty five muscle fibers were harvested from soleus (Sol) and extensor digitorum longus (EDL) muscles from TIEG1-/- (N = 5) and control (N = 5) mice. Mechanical tests were performed on fibers and the dynamic and static stresses were measured. A viscoelastic Hill model of 3rd order was used to fit the experimental relaxation test data. In parallel, immunohistochemical analyses were performed on three serial transverse sections to detect the myosin isoforms within the slow and fast muscles. The percentage and the mean cross sectional area of each fiber type were calculated. These tests revealed a significant increase in the mechanical stress properties for the TIEG1-/- Sol fibers while a significant decrease appeared for the TIEG1-/- EDL fibers. Hill model tracked the shape of the experimental relaxation curve for both genotypes and both fiber types. Immunohistochemical results showed hypertrophy of all fiber types for TIEG1-/- muscles with an increase in the percentage of glycolytic fibers (IIX, and IIB) and a decrease of oxidative fibers (I, and IIA). This study has provided new insights into the role of TIEG1, known as KLF10, in the functional (SoltypeI: more resistant, EDLtypeIIB: less resistant) and morphological (glycolytic hypertrophy) properties of fast and slow twitch skeletal muscles. Further investigation at the cellular level will better reveal the role of the TIEG1 gene in skeletal muscle tissue.
Collapse
Affiliation(s)
- Malek Kammoun
- Biomechanics and Bioengineering Laboratory, UMR CNRS 7338, Sorbonne University, Université de Technologie de Compiègne, Compiègne, France
| | - Philippe Pouletaut
- Biomechanics and Bioengineering Laboratory, UMR CNRS 7338, Sorbonne University, Université de Technologie de Compiègne, Compiègne, France
| | - Francis Canon
- Biomechanics and Bioengineering Laboratory, UMR CNRS 7338, Sorbonne University, Université de Technologie de Compiègne, Compiègne, France
| | - Malayannan Subramaniam
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota, 55905, United States of America
| | - John R. Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota, 55905, United States of America
| | - Muriel Vayssade
- Biomechanics and Bioengineering Laboratory, UMR CNRS 7338, Sorbonne University, Université de Technologie de Compiègne, Compiègne, France
| | - Sabine F. Bensamoun
- Biomechanics and Bioengineering Laboratory, UMR CNRS 7338, Sorbonne University, Université de Technologie de Compiègne, Compiègne, France
- * E-mail:
| |
Collapse
|
28
|
Kumar S, Pan CC, Shah N, Wheeler SE, Hoyt KR, Hempel N, Mythreye K, Lee NY. Activation of Mitofusin2 by Smad2-RIN1 Complex during Mitochondrial Fusion. Mol Cell 2016; 62:520-31. [PMID: 27184078 DOI: 10.1016/j.molcel.2016.04.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 02/04/2016] [Accepted: 04/07/2016] [Indexed: 01/11/2023]
Abstract
Smads are nuclear-shuttling transcriptional mediators of transforming growth factor-β (TGF-β) signaling. Although their essential nuclear roles in gene regulation during development and carcinogenesis are well established, whether they have important cytoplasmic functions remains unclear. Here we report that Smad2 is a critical determinant of mitochondrial dynamics. We identified mitofusin2 (MFN2) and Rab and Ras Interactor 1 (RIN1) as new Smad2 binding partners required for mitochondrial fusion. Unlike TGF-β-induced Smad2/3 transcriptional responses underlying mitochondrial fragmentation and apoptosis, inactive cytoplasmic Smad2 rapidly promotes mitochondrial fusion by recruiting RIN1 into a complex with MFN2. We demonstrate that Smad2 is a key scaffold, allowing RIN1 to act as a GTP exchange factor for MFN2-GTPase activation to promote mitochondrial ATP synthesis and suppress superoxide production. These results reveal functional implications between Smads and mitochondrial dysfunction in cancer and metabolic and neurodegenerative disorders.
Collapse
Affiliation(s)
- Sanjay Kumar
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Christopher C Pan
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Nirav Shah
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Sarah E Wheeler
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Kari R Hoyt
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Nadine Hempel
- Department of Pharmacology, Penn State University, Hershey, PA 17033, USA
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Nam Y Lee
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
29
|
Inhibition of Transforming Growth Factor-β Receptor signaling promotes culture expansion of undifferentiated human Endometrial Mesenchymal Stem/stromal Cells. Sci Rep 2015; 5:15042. [PMID: 26461813 PMCID: PMC4602195 DOI: 10.1038/srep15042] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Human endometrial MSC (eMSC) are a novel source of MSC easily harvested from the highly regenerative uterine lining. We have developed protocols for eMSC isolation from single cell suspensions using magnetic bead-sorting using a perivascular marker antibody to SUSD2 and culture expansion in serum free medium (SFM). Similar to other MSC, eMSC spontaneously differentiate into fibroblasts during culture expansion decreasing their purity and efficacy. The aim of this study was to determine if A83-01, a TGF-β receptor inhibitor prevents eMSC differentiation in culture. SUSD2+ eMSC were cultured in SFM with bFGF/EGF in 5% O2/5% CO2. At passage 6, eMSC were incubated with or without A83-01 for 7 days, then analysed for MSC properties. A83-01 dose dependently promoted SUSD2+ eMSC proliferation and blocked apoptosis via the SMAD 2/3 pathway. Fewer A83-01 treated cells were autofluorescent or stained with β-galactosidase, indicating reduced senescence. A83-01-treated cells had higher cloning efficiency, differentiated into mesodermal lineages and expressed MSC phenotypic markers. These data suggest that A83-01 maintains SUSD2+ eMSC stemness, promoting proliferation by blocking senescence and apoptosis in late passage cultures through binding to TGF-β receptors. Small molecules such as A83-01 may enable the expansion of undifferentiated MSC for use in tissue engineering and cell-based therapies.
Collapse
|
30
|
Jim HS, Lin HY, Tyrer JP, Lawrenson K, Dennis J, Chornokur G, Chen Z, Chen AY, Permuth-Wey J, Aben KKH, Anton-Culver H, Antonenkova N, Bruinsma F, Bandera EV, Bean YT, Beckmann MW, Bisogna M, Bjorge L, Bogdanova N, Brinton LA, Brooks-Wilson A, Bunker CH, Butzow R, Campbell IG, Carty K, Chang-Claude J, Cook LS, Cramer DW, Cunningham JM, Cybulski C, Dansonka-Mieszkowska A, du Bois A, Despierre E, Sieh W, Doherty JA, Dörk T, Dürst M, Easton DF, Eccles DM, Edwards RP, Ekici AB, Fasching PA, Fridley BL, Gao YT, Gentry-Maharaj A, Giles GG, Glasspool R, Goodman MT, Gronwald J, Harter P, Hasmad HN, Hein A, Heitz F, Hildebrandt MA, Hillemanns P, Hogdall CK, Hogdall E, Hosono S, Iversen ES, Jakubowska A, Jensen A, Ji BT, Karlan BY, Kellar M, Kiemeney LA, Krakstad C, Kjaer SK, Kupryjanczyk J, Vierkant RA, Lambrechts D, Lambrechts S, Le ND, Lee AW, Lele S, Leminen A, Lester J, Levine DA, Liang D, Lim BK, Lissowska J, Lu K, Lubinski J, Lundvall L, Massuger LF, Matsuo K, McGuire V, McLaughlin JR, McNeish I, Menon U, Milne RL, Modugno F, Thomsen L, Moysich KB, Ness RB, Nevanlinna H, Eilber U, Odunsi K, Olson SH, Orlow I, Orsulic S, et alJim HS, Lin HY, Tyrer JP, Lawrenson K, Dennis J, Chornokur G, Chen Z, Chen AY, Permuth-Wey J, Aben KKH, Anton-Culver H, Antonenkova N, Bruinsma F, Bandera EV, Bean YT, Beckmann MW, Bisogna M, Bjorge L, Bogdanova N, Brinton LA, Brooks-Wilson A, Bunker CH, Butzow R, Campbell IG, Carty K, Chang-Claude J, Cook LS, Cramer DW, Cunningham JM, Cybulski C, Dansonka-Mieszkowska A, du Bois A, Despierre E, Sieh W, Doherty JA, Dörk T, Dürst M, Easton DF, Eccles DM, Edwards RP, Ekici AB, Fasching PA, Fridley BL, Gao YT, Gentry-Maharaj A, Giles GG, Glasspool R, Goodman MT, Gronwald J, Harter P, Hasmad HN, Hein A, Heitz F, Hildebrandt MA, Hillemanns P, Hogdall CK, Hogdall E, Hosono S, Iversen ES, Jakubowska A, Jensen A, Ji BT, Karlan BY, Kellar M, Kiemeney LA, Krakstad C, Kjaer SK, Kupryjanczyk J, Vierkant RA, Lambrechts D, Lambrechts S, Le ND, Lee AW, Lele S, Leminen A, Lester J, Levine DA, Liang D, Lim BK, Lissowska J, Lu K, Lubinski J, Lundvall L, Massuger LF, Matsuo K, McGuire V, McLaughlin JR, McNeish I, Menon U, Milne RL, Modugno F, Thomsen L, Moysich KB, Ness RB, Nevanlinna H, Eilber U, Odunsi K, Olson SH, Orlow I, Orsulic S, Palmieri Weber R, Paul J, Pearce CL, Pejovic T, Pelttari LM, Pike MC, Poole EM, Schernhammer E, Risch HA, Rosen B, Rossing MA, Rothstein JH, Rudolph A, Runnebaum IB, Rzepecka IK, Salvesen HB, Schwaab I, Shu XO, Shvetsov YB, Siddiqui N, Song H, Southey MC, Spiewankiewicz B, Sucheston-Campbell L, Teo SH, Terry KL, Thompson PJ, Tangen IL, Tworoger SS, van Altena AM, Vergote I, Walsh CS, Wang-Gohrke S, Wentzensen N, Whittemore AS, Wicklund KG, Wilkens LR, Wu AH, Wu X, Woo YL, Yang H, Zheng W, Ziogas A, Amankwah E, Berchuck A, Schildkraut JM, Kelemen LE, Ramus SJ, Monteiro AN, Goode EL, Narod SA, Gayther SA, Pharoah PDP, Sellers TA, Phelan CM. Common Genetic Variation in Circadian Rhythm Genes and Risk of Epithelial Ovarian Cancer (EOC). JOURNAL OF GENETICS AND GENOME RESEARCH 2015; 2:017. [PMID: 26807442 PMCID: PMC4722961 DOI: 10.23937/2378-3648/1410017] [Show More Authors] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Disruption in circadian gene expression, whether due to genetic variation or environmental factors (e.g., light at night, shiftwork), is associated with increased incidence of breast, prostate, gastrointestinal and hematologic cancers and gliomas. Circadian genes are highly expressed in the ovaries where they regulate ovulation; circadian disruption is associated with several ovarian cancer risk factors (e.g., endometriosis). However, no studies have examined variation in germline circadian genes as predictors of ovarian cancer risk and invasiveness. The goal of the current study was to examine single nucleotide polymorphisms (SNPs) in circadian genes BMAL1, CRY2, CSNK1E, NPAS2, PER3, REV1 and TIMELESS and downstream transcription factors KLF10 and SENP3 as predictors of risk of epithelial ovarian cancer (EOC) and histopathologic subtypes. The study included a test set of 3,761 EOC cases and 2,722 controls and a validation set of 44,308 samples including 18,174 (10,316 serous) cases and 26,134 controls from 43 studies participating in the Ovarian Cancer Association Consortium (OCAC). Analysis of genotype data from 36 genotyped SNPs and 4600 imputed SNPs indicated that the most significant association was rs117104877 in BMAL1 (OR = 0.79, 95% CI = 0.68-0.90, p = 5.59 × 10-4]. Functional analysis revealed a significant down regulation of BMAL1 expression following cMYC overexpression and increasing transformation in ovarian surface epithelial (OSE) cells as well as alternative splicing of BMAL1 exons in ovarian and granulosa cells. These results suggest that variation in circadian genes, and specifically BMAL1, may be associated with risk of ovarian cancer, likely through disruption of hormonal pathways.
Collapse
Affiliation(s)
- Heather S.L. Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Hui-Yi Lin
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan P. Tyrer
- Department of Public Health and Primary Care, The Centre for Cancer Epidemiology, University of Cambridge, Strange ways Research Laboratory, Cambridge, UK
| | - Kate Lawrenson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Joe Dennis
- Department of Public Health and Primary Care, The Centre for Cancer Epidemiology, University of Cambridge, Strange ways Research Laboratory, Cambridge, UK
| | - Ganna Chornokur
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA
| | - Zhihua Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Ann Y. Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Jennifer Permuth-Wey
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA
| | - Katja KH. Aben
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands
| | - Hoda Anton-Culver
- Genetic Epidemiology Research Institute, UCI Center for Cancer Genetics Research and Prevention, School of Medicine, Department of Epidemiology, University of California Irvine, Irvine, CA, USA
| | - Natalia Antonenkova
- Byelorussian Institute for Oncology and Medical Radiology Aleksandrov N.N., Minsk, Belarus
| | - Fiona Bruinsma
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
| | - Elisa V. Bandera
- Cancer Prevention and Control, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Yukie T. Bean
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University, Erlangen-Nuremberg Comprehensive Cancer Center, Erlangen EMN, Germany
| | - Maria Bisogna
- Department of Surgery, Gynecology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Line Bjorge
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Natalia Bogdanova
- Gynecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Louise A. Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Angela Brooks-Wilson
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC Canada
| | - Clareann H. Bunker
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Ralf Butzow
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, HUS, Finland
- Department of Pathology, Helsinki University Central Hospital, Helsinki, HUS, Finland
| | - Ian G. Campbell
- Cancer Genetics Laboratory, Research Division, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Karen Carty
- Department of Gynaecological Oncology, Glasgow Royal Infirmary, Glasgow, G31 2ER, UK
- CRUK Clinical Trials Unit, The Beatson West of Scotland Cancer Centre, 1053 Great Western Road, Glasgow G12 0YN, UK
| | - Jenny Chang-Claude
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Linda S. Cook
- Division of Epidemiology and Biostatistics, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Daniel W. Cramer
- Obstetrics and Gynecology Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie M. Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | | | - Andreas du Bois
- Department of Gynaecology and Gynaecologic Oncology, Kliniken Essen-Mitte/ Evang. Huyssens-Stiftung/Knappschaft GmbH, Essen, Germany
- Department of Gynaecology and Gynaecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | - Evelyn Despierre
- Division of Gynecologic Oncology; Leuven Cancer Institute, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Weiva Sieh
- Department of Health Research and Policy-Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer A. Doherty
- Department of Epidemiology, Geisel School of Medicine, Dartmouth, Hanover, NH, USA
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Thilo Dörk
- Gynecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Matthias Dürst
- Department of Gynecology, Friedrich Schiller University, Jena, Germany
| | - Douglas F. Easton
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Diana M. Eccles
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Robert P. Edwards
- Department of Obstetrics Gynecology/RS, Division of Gynecological Oncology, Ovarian Cancer Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Arif B. Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University, Erlangen-Nuremberg Comprehensive Cancer Center, Erlangen EMN, Germany
- Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Brooke L. Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | | | - Graham G. Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Rosalind Glasspool
- CRUK Clinical Trials Unit, The Beatson West of Scotland Cancer Centre, 1053 Great Western Road, Glasgow G12 0YN, UK
| | - Marc T. Goodman
- Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Community and Population Health Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Philipp Harter
- Department of Gynaecology and Gynaecologic Oncology, Kliniken Essen-Mitte/ Evang. Huyssens-Stiftung/Knappschaft GmbH, Essen, Germany
- Department of Gynaecology and Gynaecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | - Hanis N. Hasmad
- Cancer Research Initiatives Foundation, Sime Darby Medical Center, Subang Jaya, Malaysia
| | - Alexander Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University, Erlangen-Nuremberg Comprehensive Cancer Center, Erlangen EMN, Germany
| | - Florian Heitz
- Department of Gynaecology and Gynaecologic Oncology, Kliniken Essen-Mitte/ Evang. Huyssens-Stiftung/Knappschaft GmbH, Essen, Germany
- Department of Gynaecology and Gynaecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | | | - Peter Hillemanns
- Gynecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Claus K. Hogdall
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Hogdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pathology, Molecular Unit, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Satoyo Hosono
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | | | - Anna Jakubowska
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Bu-Tian Ji
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Beth Y. Karlan
- Women’s Cancer Program at the Samuel Oschin Comprehensive, Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Melissa Kellar
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Lambertus A. Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Camilla Krakstad
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Susanne K. Kjaer
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Jolanta Kupryjanczyk
- Department of Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Robert A. Vierkant
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Diether Lambrechts
- Vesalius Research Center, VIB, University of Leuven, Leuven, Belgium
- Department of Oncology, Laboratory for Translational Genetics, University of Leuven, Belgium
| | - Sandrina Lambrechts
- Division of Gynecologic Oncology; Leuven Cancer Institute, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Nhu D. Le
- Cancer Control Research, BC Cancer Agency, Vancouver, BC, Canada
| | - Alice W. Lee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Shashi Lele
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Arto Leminen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, HUS, Finland
| | - Jenny Lester
- Women’s Cancer Program at the Samuel Oschin Comprehensive, Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Douglas A. Levine
- Department of Surgery, Gynecology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Dong Liang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Boon Kiong Lim
- Department of Obstetrics and Gynaecology, University Malaya Medical Centre, University Malaya, Kuala Lumpur, Malaysia
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Karen Lu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jan Lubinski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Lene Lundvall
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Leon F.A.G. Massuger
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Keitaro Matsuo
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Valerie McGuire
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ian McNeish
- CRUK Clinical Trials Unit, The Beatson West of Scotland Cancer Centre, 1053 Great Western Road, Glasgow G12 0YN, UK
| | - Usha Menon
- Women’s Cancer, UCL EGA Institute for Women’s Health, London, UK
| | - Roger L. Milne
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Francesmary Modugno
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Women’s Cancer Research Program, Magee-Women’s Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lotte Thomsen
- Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Roberta B. Ness
- The University of Texas School of Public Health, Houston, TX, USA
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, HUS, Finland
| | - Ursula Eilber
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY
| | - Sara H. Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Sandra Orsulic
- Women’s Cancer Program at the Samuel Oschin Comprehensive, Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rachel Palmieri Weber
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - James Paul
- CRUK Clinical Trials Unit, The Beatson West of Scotland Cancer Centre, 1053 Great Western Road, Glasgow G12 0YN, UK
| | - Celeste L. Pearce
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
- Department of Epidemiology, University of Michigan, 1415 Washington Heights, Ann Arbor, Michigan, USA
| | - Tanja Pejovic
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Liisa M. Pelttari
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, HUS, Finland
| | - Malcolm C. Pike
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Elizabeth M. Poole
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Eva Schernhammer
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Harvey A. Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Barry Rosen
- Department of Gynecology-Oncology, Princess Margaret Hospital, and Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Joseph H. Rothstein
- Department of Health Research and Policy-Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anja Rudolph
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Ingo B. Runnebaum
- Department of Gynecology, Friedrich Schiller University, Jena, Germany
| | - Iwona K. Rzepecka
- Department of Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Helga B. Salvesen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ira Schwaab
- Institut für Humangenetik, Wiesbaden, Germany
| | - Xiao-Ou Shu
- Epidemiology Center and Vanderbilt, Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yurii B. Shvetsov
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Nadeem Siddiqui
- Department of Gynaecological Oncology, Glasgow Royal Infirmary, Glasgow, G31 2ER, UK
| | - Honglin Song
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Melissa C. Southey
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | | | - Lara Sucheston-Campbell
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Soo-Hwang Teo
- Cancer Research Initiatives Foundation, Sime Darby Medical Center, Subang Jaya, Malaysia
- University Malaya Medical Centre, University of Malaya, Kuala Lumpur, Maylaysia
| | - Kathryn L. Terry
- Obstetrics and Gynecology Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Pamela J. Thompson
- Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Community and Population Health Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ingvild L. Tangen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Shelley S. Tworoger
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Anne M. van Altena
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Ignace Vergote
- Division of Gynecologic Oncology; Leuven Cancer Institute, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Christine S. Walsh
- Women’s Cancer Program at the Samuel Oschin Comprehensive, Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shan Wang-Gohrke
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alice S. Whittemore
- Department of Health Research and Policy-Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristine G. Wicklund
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Lynne R. Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Anna H. Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yin-Ling Woo
- Department of Obstetrics and Gynaecology, University Malaya Medical Centre, University Malaya, Kuala Lumpur, Malaysia
| | - Hannah Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zheng
- Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Argyrios Ziogas
- Genetic Epidemiology Research Institute, UCI Center for Cancer Genetics Research and Prevention, School of Medicine, Department of Epidemiology, University of California Irvine, Irvine, CA, USA
| | - Ernest Amankwah
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA
- Clinical and Translational Research Organization, All Children’s Hospital Johns Hopkins Medicine, St Petersburg, FL
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | | | - Joellen M. Schildkraut
- Cancer Prevention, Detection & Control Research Program, Duke Cancer Institute, Durham, NC, USA
| | - Linda E. Kelemen
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Susan J. Ramus
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Alvaro N.A. Monteiro
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA
| | - Ellen L. Goode
- Department of Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Steven A. Narod
- Women’s College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Simon A. Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Paul D. P. Pharoah
- Department of Public Health and Primary Care, The Centre for Cancer Epidemiology, University of Cambridge, Strange ways Research Laboratory, Cambridge, UK
- The Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Thomas A. Sellers
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA
| | - Catherine M. Phelan
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
31
|
Klf10 regulates odontoblast differentiation and mineralization via promoting expression of dentin matrix protein 1 and dentin sialophosphoprotein genes. Cell Tissue Res 2015; 363:385-98. [PMID: 26310138 DOI: 10.1007/s00441-015-2260-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 07/28/2015] [Indexed: 01/31/2023]
Abstract
Klf10, a member of the Krüppel-like family of transcription factors, is critical for osteoblast differentiation, bone formation and mineralization. However, whether Klf10 is involved in odontoblastic differentiation and tooth development has not been determined. In this study, we investigate the expression patterns of Klf10 during murine tooth development in vivo and its role in odontoblastic differentiation in vitro. Klf10 protein was expressed in the enamel organ and the underlying mesenchyme, ameloblasts and odontoblasts at early and later stages of murine molar formation. Furthermore, the expression of Klf10, Dmp1, Dspp and Runx2 was significantly elevated during the process of mouse dental papilla mesenchymal differentiation and mineralization. The overexpression of Klf10 induced dental papilla mesenchymal cell differentiation and mineralization as detected by alkaline phosphatase staining and alizarin red S assay. Klf10 additionally up-regulated the expression of odontoblastic differentiation marker genes Dmp1, Dspp and Runx2 in mouse dental papilla mesenchymal cells. The molecular mechanism of Klf10 in controlling Dmp1 and Dspp expression is thus to activate their regulatory regions in a dosage-dependent manner. Our results suggest that Klf10 is involved in tooth development and promotes odontoblastic differentiation via the up-regulation of Dmp1 and Dspp transcription.
Collapse
|
32
|
Human cancer: Is it linked to dysfunctional lipid metabolism? Biochim Biophys Acta Gen Subj 2015; 1850:352-64. [DOI: 10.1016/j.bbagen.2014.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 11/23/2022]
|
33
|
Martínez-Armenta M, Díaz de León-Guerrero S, Catalán A, Alvarez-Arellano L, Uribe RM, Subramaniam M, Charli JL, Pérez-Martínez L. TGFβ2 regulates hypothalamic Trh expression through the TGFβ inducible early gene-1 (TIEG1) during fetal development. Mol Cell Endocrinol 2015; 400:129-39. [PMID: 25448845 PMCID: PMC4415168 DOI: 10.1016/j.mce.2014.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 09/01/2014] [Accepted: 10/27/2014] [Indexed: 01/05/2023]
Abstract
The hypothalamus regulates the homeostasis of the organism by controlling hormone secretion from the pituitary. The molecular mechanisms that regulate the differentiation of the hypothalamic thyrotropin-releasing hormone (TRH) phenotype are poorly understood. We have previously shown that Klf10 or TGFβ inducible early gene-1 (TIEG1) is enriched in fetal hypothalamic TRH neurons. Here, we show that expression of TGFβ isoforms (1-3) and both TGFβ receptors (TβRI and II) occurs in the hypothalamus concomitantly with the establishment of TRH neurons during late embryonic development. TGFβ2 induces Trh expression via a TIEG1 dependent mechanism. TIEG1 regulates Trh expression through an evolutionary conserved GC rich sequence on the Trh promoter. Finally, in mice deficient in TIEG1, Trh expression is lower than in wild type animals at embryonic day 17. These results indicate that TGFβ signaling, through the upregulation of TIEG1, plays an important role in the establishment of Trh expression in the embryonic hypothalamus.
Collapse
MESH Headings
- Animals
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Embryo, Mammalian
- Fetus
- Gene Expression Regulation, Developmental
- Hypothalamus/cytology
- Hypothalamus/growth & development
- Hypothalamus/metabolism
- Immunohistochemistry
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons/cytology
- Neurons/metabolism
- Primary Cell Culture
- Promoter Regions, Genetic
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Rats
- Rats, Wistar
- Receptor, Transforming Growth Factor-beta Type I
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction
- Thyrotropin-Releasing Hormone/genetics
- Thyrotropin-Releasing Hormone/metabolism
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta2/genetics
- Transforming Growth Factor beta2/metabolism
- Transforming Growth Factor beta3/genetics
- Transforming Growth Factor beta3/metabolism
Collapse
Affiliation(s)
- Miriam Martínez-Armenta
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, Mexico
| | - Sol Díaz de León-Guerrero
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, Mexico
| | - Ana Catalán
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, Mexico
| | - Lourdes Alvarez-Arellano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, Mexico
| | - Rosa Maria Uribe
- Departamento de Genética y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Morelos, Mexico
| | | | - Jean-Louis Charli
- Departamento de Genética y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Morelos, Mexico
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, Mexico.
| |
Collapse
|
34
|
Han Y, Jiang Q, Gao H, Fan J, Wang Z, Zhong F, Zheng Y, Gong Z, Wang C. The Anti-apoptotic Effect of Polypeptide from Chlamys farreri (PCF) in UVB-Exposed HaCaT Cells Involves Inhibition of iNOS and TGF-β1. Cell Biochem Biophys 2014; 71:1105-15. [DOI: 10.1007/s12013-014-0315-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
35
|
Hawse JR, Pitel KS, Cicek M, Philbrick KA, Gingery A, Peters KD, Syed FA, Ingle JN, Suman VJ, Iwaniec UT, Turner RT, Spelsberg TC, Subramaniam M. TGFβ inducible early gene-1 plays an important role in mediating estrogen signaling in the skeleton. J Bone Miner Res 2014; 29:1206-16. [PMID: 24190163 PMCID: PMC4028712 DOI: 10.1002/jbmr.2142] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 10/14/2013] [Accepted: 10/27/2013] [Indexed: 12/26/2022]
Abstract
TGFβ Inducible Early Gene-1 (TIEG1) knockout (KO) mice display a sex-specific osteopenic phenotype characterized by low bone mineral density, bone mineral content, and overall loss of bone strength in female mice. We, therefore, speculated that loss of TIEG1 expression would impair the actions of estrogen on bone in female mice. To test this hypothesis, we employed an ovariectomy (OVX) and estrogen replacement model system to comprehensively analyze the role of TIEG1 in mediating estrogen signaling in bone at the tissue, cell, and biochemical level. Dual-energy X-ray absorptiometry (DXA), peripheral quantitative computed tomography (pQCT), and micro-CT analyses revealed that loss of TIEG1 expression diminished the effects of estrogen throughout the skeleton and within multiple bone compartments. Estrogen exposure also led to reductions in bone formation rates and mineralizing perimeter in wild-type mice with little to no effects on these parameters in TIEG1 KO mice. Osteoclast perimeter per bone perimeter and resorptive activity as determined by serum levels of CTX-1 were differentially regulated after estrogen treatment in TIEG1 KO mice compared with wild-type littermates. No significant differences were detected in serum levels of P1NP between wild-type and TIEG1 KO mice. Taken together, these data implicate an important role for TIEG1 in mediating estrogen signaling throughout the mouse skeleton and suggest that defects in this pathway are likely to contribute to the sex-specific osteopenic phenotype observed in female TIEG1 KO mice.
Collapse
Affiliation(s)
- John R. Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Kevin S. Pitel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Muzaffer Cicek
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth A. Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Anne Gingery
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth D. Peters
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Farhan A. Syed
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | | | - Vera J. Suman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Thomas C. Spelsberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
36
|
Hwang YC, Yang CH, Lin CH, Ch'ang HJ, Chang VHS, Yu WCY. Destabilization of KLF10, a tumor suppressor, relies on thr93 phosphorylation and isomerase association. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3035-3045. [PMID: 23994618 DOI: 10.1016/j.bbamcr.2013.08.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 08/08/2013] [Accepted: 08/12/2013] [Indexed: 12/24/2022]
Abstract
KLF10 is now classified as a member of the Krüppel-like transcription factor family and acts as a tumor suppressor. Although KLF10 is originally named as TGF-β-inducible early gene-1 and mimicking the anti-proliferative effect of TGF-β in various carcinoma cells, the transcriptional upregulatory function of KLF10 has been described for a variety of cytokines and in many diseases. Through in vivo and in vitro phosphorylation assays, we identified that KLF10 is a phosphorylated protein in cells. Using yeast-two hybrid screening and site direct mutagenesis, we also identified PIN1 as a novel KLF10 associated protein. PIN1 is a peptidyl-prolyl isomerase enzyme belonging to the parvulin family, which specifically recognizes phosphorylated Ser/Thr-Pro containing substrates. Through protein-protein interaction assays, we showed that the Pro-directed Ser/Thr-Pro motif at Thr-93 in the KLF10 N-terminal region is essential for the interaction between KLF10 and PIN1. More importantly, PIN1 interacts with KLF10 in a phosphorylation-dependent manner and this interaction promotes KLF10 protein degradation in cells. Therefore, KLF10 shows shorter protein stability compared with mutant KLF10 that lacks PIN1 binding ability after cycloheximide treatments. The reversely correlated expression profile between KLF10 and PIN1 as observed in cell lines was also shown in clinic pancreatic cancer specimen. Using in vitro kinase assays and depletion assays, we were able to show that RAF-1 phosphorylates the Thr-93 of KLF10 and affects the KLF10 expression level in cells. Thus these findings as a whole indicate that RAF-1 phosphorylation and PIN1 isomerization together regulate KLF10 stability and further affect the role of KLF10 in tumor progression.
Collapse
Affiliation(s)
- Yu-Chyi Hwang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chien-Hui Yang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Ching-Hui Lin
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-Ju Ch'ang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Vincent H S Chang
- Program for Translation Medicine, College of Medical Science and Technology, Taipei Medical University, Taiwan.
| | - Winston C Y Yu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
37
|
Gentle ME, Shi S, Daehn I, Zhang T, Qi H, Yu L, D'Agati VD, Schlondorff DO, Bottinger EP. Epithelial cell TGFβ signaling induces acute tubular injury and interstitial inflammation. J Am Soc Nephrol 2013; 24:787-99. [PMID: 23539761 DOI: 10.1681/asn.2012101024] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
TGFβ signaling plays a central role in the development of acute and chronic kidney diseases. Previous in vivo studies involved systemic alteration of TGFβ signaling, however, limiting conclusions about the direct role of TGFβ in tubular cell injury. Here, we generated a double transgenic mouse that inducibly expresses a ligand-independent constitutively active TGFβ receptor type 1 (TβR1) kinase specifically in tubular epithelial cells, with expression restricted by the Pax8 promoter. In this model, activation of TGFβ signaling in the tubular epithelium alone was sufficient to cause AKI characterized by marked tubular cell apoptosis and necrosis, oxidative stress, dedifferentiation and regenerative cell proliferation, reduced renal function, and interstitial accumulation of inflammatory cells. This tubular injury was associated with mitochondrial-derived generation of reactive oxygen species (ROS), but cell damage and apoptosis were partially independent of mitochondrial-derived ROS. TβR1 signaling-induced tubular injury also associated with significant leukocyte infiltration consisting of F4/80(+) macrophages, CD11c(+) F4/80(+) dendritic cells, CD11c(+) F4/80(-) Ly6C(high) dendritic cells/monocytes, and T cells. Inhibition of mitochondrial-derived ROS significantly reduced accumulation of CD11c(+) F4/80(+) dendritic cells and T cells, suggesting a role for ROS in the activation and recruitment of the adaptive immune response to tubular injury. Taken together, these results suggest that TGFβ signaling in the tubular epithelium alone is sufficient to cause acute tubular injury and inflammation; therefore, TGFβ may be a mechanistic link between acute injury and chronic progression of kidney disease.
Collapse
Affiliation(s)
- Madeleine E Gentle
- Division of Nephrology, Department of Medicine, Mount Sinai School of Medicine, One Gustave Levy Place, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Temme J, Bauer G. Low-dose gamma irradiation enhances superoxide anion production by nonirradiated cells through TGF-β1-dependent bystander signaling. Radiat Res 2013; 179:422-32. [PMID: 23465059 DOI: 10.1667/rr3161.2] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We show here that low-dose gamma irradiation substantially increase in extracellular superoxide anion production in oncogenically transformed cells and tumor cells but not by nontransformed cells. The transfer of only a few cells from an irradiated culture to nonirradiated control cells was sufficient for the transmission of a signal to induce superoxide anion production in the nonirradiated cells. The number of irradiated cells that was necessary for the successful induction of superoxide anion production in the nonirradiated cells depended on radiation dose. When irradiated cells were allowed to incubate for 1 h before transmission to the nonirradiated cultures, nearly all of the cells from the irradiated cell population were able to communicate the inducing signal to nonirradiated cells. siRNA-dependent knockdown and reconstitution experiments showed that TGF-β1 was sufficient to mediate the bystander effect triggered by low-dose radiation in this experimental system. A kinetic analysis demonstrated that the enhanced superoxide anion production was substantially reduced before the release of the bystander signal by activated TGF-β.
Collapse
Affiliation(s)
- Jennifer Temme
- Abteilung Virologie, Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, Germany
| | | |
Collapse
|
39
|
Casalena G, Daehn I, Bottinger E. Transforming growth factor-β, bioenergetics, and mitochondria in renal disease. Semin Nephrol 2012; 32:295-303. [PMID: 22835461 DOI: 10.1016/j.semnephrol.2012.04.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The transforming growth factor-β (TGF-β) family comprises more than 30 family members that are structurally related secreted dimeric cytokines, including TGF-β, activins, and bone morphogenetic proteins/growth and differentiation factors. TGF-β are pluripotent regulators of cell proliferation, differentiation, apoptosis, migration, and adhesion of many different cell types. TGF-β pathways are highly evolutionarily conserved and control embryogenesis, tissue repair, and tissue homeostasis in invertebrates and vertebrates. Aberrations in TGF-β activity and signaling underlie a broad spectrum of developmental disorders and major pathologies in human beings, including cancer, fibrosis, and autoimmune diseases. Recent observations have indicated an emerging role for TGF-β in the regulation of mitochondrial bioenergetics and oxidative stress responses characteristic of chronic degenerative diseases and aging. Conversely, energy and metabolic sensory pathways cross-regulate mediators of TGF-β signaling. Here, we review TGF-β and regulation of bioenergetic and mitochondrial functions, including energy and oxidant metabolism and apoptotic cell death, as well as their emerging relevance in renal biology and disease.
Collapse
Affiliation(s)
- Gabriella Casalena
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
40
|
Krüppel-like factor 10 expression as a prognostic indicator for pancreatic adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:423-30. [PMID: 22688058 DOI: 10.1016/j.ajpath.2012.04.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 04/19/2012] [Accepted: 04/24/2012] [Indexed: 12/14/2022]
Abstract
Deregulation of transforming growth factor (TGF)-β function is a common feature of pancreatic cancer, rendering these cancers unresponsive to TGF-β-stimulated growth inhibition. Recent findings have supported a primary role for Krüppel-like factor 10 (KLF10) as an important transcription factor involved in mediating TGF-β1 signaling. The aim of this study was to evaluate the correlation between KLF10 expression and the clinical and pathologic features of pancreatic cancer. Tissue specimens from patients with pancreatic adenocarcinoma were retrospectively collected for immunohistochemical analysis. To demonstrate that Klf10 expression was primarily regulated by methylation status, the Klf10 promoter was examined by methylation-specific PCR using a pancreatic cancer cell line (Panc-1). DNA methyltransferase (DNMT) inhibitor and small-interfering RNA depletion of DNMT genes were used to reverse KLF10 expression in the Panc-1 cells. In parallel, DNMT1 expression was evaluated in the pancreatic cancer tissue specimens. In 95 pancreatic cancer tissue specimens, KLF10 expression was inversely correlated with pancreatic cancer stage (P = 0.01). Multivariable analysis revealed that, in addition to the presence of distant metastasis at diagnosis (P = 0.001 and 0.001, respectively), KLF10 was another independent prognostic factor related to progression-free and overall survival (P = 0.018 and 0.037, respectively). The loss of KLF10 expression in advanced pancreatic cancer is correlated with altered methylation status, which seems to be regulated by DNMT1. Our results suggest that KLF10 is a potential clinical predictor for progression of pancreatic cancer.
Collapse
|
41
|
Jiang L, Lai YK, Zhang JF, Chan CY, Lu G, Lin MCM, He ML, Li JC, Kung HF. Transactivation of the TIEG1 confers growth inhibition of transforming growth factor-β-susceptible hepatocellular carcinoma cells. World J Gastroenterol 2012; 18:2035-2042. [PMID: 22563190 PMCID: PMC3342601 DOI: 10.3748/wjg.v18.i17.2035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 11/12/2011] [Accepted: 02/26/2012] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the role of transforming growth factor (TGF)-β-inducible early gene 1 (TIEG1) in TGF-β-induced growth inhibition in hepatocellular carcinoma (HCC) cells. METHODS Human hepatocyte and HCC cell lines with varied susceptibilities to TGF-β1 were tested by methylthiazoletetrazolium (MTT) assay. The expression changes of Smad2, Smad3, Smad4, Smad7, TIEG1 and TIEG2 gene following treatment with TGF-β1 in a TGF-β-sensitive hepatocyte cell line (MIHA), a TGF-β-sensitive hepatoma cell line (Hep3B) and two TGF-β-insensitive hepatoma cell lines (HepG2 and Bel7404) were examined. SiRNA targeting TIEG1 was transfected into Hep3B cells and the sensitivity of cells to TGF-β1 was examined. Overexpression of TIEG1 was induced by lentiviral-mediated transduction in TGF-β1-resistant hepatoma cell lines (Bel7404 and HepG2). MTT assay and 4',6-Diamidino-2-phenylindole staining were used to identify cell viability and apoptosis, respectively. The expression level of stathmin was measured by reverse transcriptase polymerase chain reaction and Western-blotting analysis, and stathmin promoter activity by TIEG1 was monitored by a luciferase reporter gene system. RESULTS TIEG1 was significantly upregulated by TGF-β1 in the TGF-β1-sensitive HCC cell line, Hep3B, but not in the resistant cell lines. The suppression of TIEG1 by siRNAs decreased the sensitivity of Hep3B cells to TGF-β1, whereas the overexpression of TIEG1 mediated growth inhibition and apoptosis in TGF-β1-resistant HCC cell lines, which resembled those of TGF-β1-sensitive HCC cells treated with TGF-β1. Our data further suggested that stathmin was a direct target of TIEG1, as stathmin was significantly downregulated by TIEG1 overexpression, and stathmin promoter activity was inhibited by TIEG1 in a dose-dependent manner. CONCLUSION Our data suggest that transactivation of TIEG1 conferred growth inhibition of TGF-β-susceptible human HCC cells.
Collapse
|
42
|
Imamura T, Hikita A, Inoue Y. The roles of TGF-β signaling in carcinogenesis and breast cancer metastasis. Breast Cancer 2011; 19:118-24. [PMID: 22139728 DOI: 10.1007/s12282-011-0321-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 11/06/2011] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-β (TGF-β) ligand is a multifunctional growth factor that regulates various cell behavior, such as cell proliferation, differentiation, migration, and apoptosis. Because TGF-β is a potent growth inhibitor, abnormalities in TGF-β signaling result in carcinogenesis. In addition to tumor suppressor function, TGF-β acts as an oncogenic factor. In particular, TGF-β signaling plays an important role during metastasis of breast cancer. Recently, epithelial-mesenchymal transition (EMT) has been shown to confer malignant properties such as cell motility and invasiveness to cancer cells and plays crucial roles during cancer metastasis. Moreover, breast stem-like cells exhibit EMT properties. Because TGF-β is a potent regulator of EMT as well as cell stemness, TGF-β signaling might play a crucial role in the regulation of breast cancer stem cells.
Collapse
Affiliation(s)
- Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan.
| | | | | |
Collapse
|
43
|
Taguchi M, Moran SL, Zobitz ME, Zhao C, Subramaniam M, Spelsberg TC, Amadio PC. WOUND-HEALING PROPERTIES OF TRANSFORMING GROWTH FACTOR β (TGF-β) INDUCIBLE EARLY GENE 1 (TIEG1) KNOCKOUT MICE. ACTA ACUST UNITED AC 2011; 11:63-69. [PMID: 20016760 DOI: 10.1142/s0218957708002012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Transforming growth factor beta (TGF-β) has a broad effect on wound healing, but many questions remain about the regulation of TGF-β during the healing process. TGF-β inducible early gene 1 (TIEG1) is a primary response gene for TGF-β that controls the activities of the TGF-β/Smad pathway, the primary TGF-β signaling pathway. The purpose of this study was to investigate the role of TIEG1 in cutaneous wound healing using TIEG1 knockout mice. The wound healing in TIEG1 knockout mice and wild-type controls was evaluated by wound breaking strength, Western blot, and histology at postoperative days 3, 7, and 14. Although re-epithelialization of both groups was similarly complete at day 7, the TIEG1 knockout mice had a significantly lower wound breaking strength than the controls at postoperative day 14. These results suggest that TIEG1 expression may be an important factor involved in the initiation and support of normal cutaneous wound healing.
Collapse
Affiliation(s)
- Manabu Taguchi
- Orthopedic Biomechanical Laboratory, Mayo Clinic 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
TIEG1 inhibits breast cancer invasion and metastasis by inhibition of epidermal growth factor receptor (EGFR) transcription and the EGFR signaling pathway. Mol Cell Biol 2011; 32:50-63. [PMID: 22025675 DOI: 10.1128/mcb.06152-11] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
TIEG1 can induce apoptosis of cancer cells, but its role in inhibiting invasion and metastasis has not been reported and is unclear. In this study, we find that decreased TIEG1 expression is associated with increased human epidermal growth factor receptor (EGFR) expression in breast cancer tissues and cell lines. TIEG1 plays an important role in suppressing transcription of EGFR by directly binding to the EGFR promoter. While overexpression of TIEG1 attenuates EGFR expression, knockdown of TIEG1 stimulates EGFR expression. Furthermore, TIEG1 and HDAC1 form a complex, which binds to Sp1 sites on the EGFR promoter and inhibits its transcription by suppressing histone acetylation. TIEG1 significantly inhibits breast cancer cell invasion, suppresses mammary tumorigenesis in xenografts in mice, and decreases lung metastasis by inhibition of EGFR gene transcription and the EGFR signaling pathway. Therefore, TIEG1 is an antimetastasis gene product; regulation of EGFR expression by TIEG1 may be part of an integral signaling pathway that determines and explains breast cancer invasion and metastasis.
Collapse
|
45
|
Cazanave SC, Mott JL, Bronk SF, Werneburg NW, Fingas CD, Meng XW, Finnberg N, El-Deiry WS, Kaufmann SH, Gores GJ. Death receptor 5 signaling promotes hepatocyte lipoapoptosis. J Biol Chem 2011; 286:39336-48. [PMID: 21941003 DOI: 10.1074/jbc.m111.280420] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nonalcoholic steatohepatitis is characterized by hepatic steatosis, elevated levels of circulating free fatty acids (FFA), endoplasmic reticulum (ER) stress, and hepatocyte lipoapoptosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor 5 (DR5) is significantly elevated in patients with nonalcoholic steatohepatitis, and steatotic hepatocytes demonstrate increased sensitivity to TRAIL-mediated cell death. Nonetheless, a role for TRAIL and/or DR5 in mediating lipoapoptotic pathways is unexplored. Here, we examined the contribution of DR5 death signaling to lipoapoptosis by free fatty acids. The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. Lipoapoptosis by palmitate was not inhibited by a soluble human recombinant DR5-Fc chimera protein suggesting that DR5 cytotoxic signaling is ligand-independent. Hepatocytes from murine TRAIL receptor knock-out mice (DR(-/-)) displayed reduced palmitate-mediated lipotoxicity. Likewise, knockdown of DR5 or caspase-8 expression by shRNA technology attenuated palmitate-induced Bax activation and apoptosis in Huh-7 cells, without altering induction of ER stress markers. Similar observations were verified in other cell models. Finally, knockdown of CHOP, an ER stress-mediated transcription factor, reduced DR5 up-regulation and DR5-mediated caspase-8 activation upon palmitate treatment. Collectively, these results suggest that ER stress-induced CHOP activation by palmitate transcriptionally up-regulates DR5, likely resulting in ligand-independent cytotoxic signaling by this death receptor.
Collapse
Affiliation(s)
- Sophie C Cazanave
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Jiang L, Wang F, Lin F, Gao SM, Tan Y, Han Y, Chen C, Wu J. Lentivirus-mediated overexpression of TGF-β inducible early gene 1 inhibits SW1990 pancreatic cancer cell growth. Cell Biol Int 2011; 35:891-896. [PMID: 21524276 DOI: 10.1042/cbi20100896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
TIEG1 (TGF-β inducible early gene 1) plays a significant role in regulating cell proliferation and apoptosis in various cell types. Previous studies have shown a close relationship between the expression level of TIEG1 and various cancers, including breast, prostate, colorectal and pancreatic cancer. In this study, we up-regulated the gene expression of TIEG1 in SW1990 pancreatic cancer cell line by a lentivirus transfection system and investigated its potential as a therapeutic target for pancreatic cancer. The results showed that lentivirus-mediated overexpression of TIEG1 gene inhibited human pancreatic cancer SW1990 cell proliferation and caused the cell cycle arrest at the G1-phase in vitro. SW1990 cells transduced with lenti-TIEG1 showed significant inhibition of colony formation and cancer cell growth in 3-D culture model. Moreover, overexpression of TIEG1 gene significantly slowed the growth of SW1990 xenografts in nude mice. Taken together, these data provided evidence that overexpression of TIEG1 gene by a lentivirus transfection system led to suppressed human pancreatic cancer cell growth and might therefore be a feasible approach in the clinical management of pancreatic cancer.
Collapse
Affiliation(s)
- Lei Jiang
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, Peoples Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Klf10 and Klf11 as mediators of TGF-beta superfamily signaling. Cell Tissue Res 2011; 347:65-72. [PMID: 21574058 DOI: 10.1007/s00441-011-1186-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/27/2011] [Indexed: 12/19/2022]
Abstract
Klf10 and Klf11 belong to the family of Sp1/Krüppel-like zinc finger transcription factors that play important roles in a variety of cell types and tissues. Although Klf10 and Klf11 were initially introduced as transforming growth factor-beta (TGF-beta)-inducible genes, several studies have described their upregulation by a plethora of growth factors, cytokines and hormones. Here, we review the current knowledge of the inductive cues for Klf10 and Klf11 and focus on their transcriptional regulation by members of the TGF-beta superfamily. We further summarize their involvement in the regulation of the TGF-beta signaling pathway and discuss their possible role as molecules mediating crosstalk between various signaling pathways. Finally, we provide an overview of the pro-apoptotic and anti-proliferative functions of Klf10 and Klf11.
Collapse
|
48
|
TGF-β inducible early gene 1 regulates osteoclast differentiation and survival by mediating the NFATc1, AKT, and MEK/ERK signaling pathways. PLoS One 2011; 6:e17522. [PMID: 21423731 PMCID: PMC3056664 DOI: 10.1371/journal.pone.0017522] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 02/04/2011] [Indexed: 01/18/2023] Open
Abstract
TGF-β Inducible Early Gene-1 (TIEG1) is a Krüppel-like transcription factor (KLF10) that was originally cloned from human osteoblasts as an early response gene to TGF-β treatment. As reported previously, TIEG1(-/-) mice have decreased cortical bone thickness and vertebral bone volume and have increased spacing between the trabeculae in the femoral head relative to wildtype controls. Here, we have investigated the role of TIEG1 in osteoclasts to further determine their potential role in mediating this phenotype. We have found that TIEG1(-/-) osteoclast precursors differentiated more slowly compared to wildtype precursors in vitro and high RANKL doses are able to overcome this defect. We also discovered that TIEG1(-/-) precursors exhibit defective RANKL-induced phosphorylation and accumulation of NFATc1 and the NFATc1 target gene DC-STAMP. Higher RANKL concentrations reversed defective NFATc1 signaling and restored differentiation. After differentiation, wildtype osteoclasts underwent apoptosis more quickly than TIEG1(-/-) osteoclasts. We observed increased AKT and MEK/ERK signaling pathway activation in TIEG1(-/-) osteoclasts, consistent with the roles of these kinases in promoting osteoclast survival. Adenoviral delivery of TIEG1 (AdTIEG1) to TIEG1(-/-) cells reversed the RANKL-induced NFATc1 signaling defect in TIEG1(-/-) precursors and eliminated the differentiation and apoptosis defects. Suppression of TIEG1 with siRNA in wildtype cells reduced differentiation and NFATc1 activation. Together, these data provide evidence that TIEG1 controls osteoclast differentiation by reducing NFATc1 pathway activation and reduces osteoclast survival by suppressing AKT and MEK/ERK signaling.
Collapse
|
49
|
Alemu EA, Sjøttem E, Outzen H, Larsen KB, Holm T, Bjørkøy G, Johansen T. Transforming growth factor-β-inducible early response gene 1 is a novel substrate for atypical protein kinase Cs. Cell Mol Life Sci 2010; 68:1953-68. [PMID: 20953893 PMCID: PMC3092057 DOI: 10.1007/s00018-010-0541-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 09/03/2010] [Accepted: 09/27/2010] [Indexed: 11/25/2022]
Abstract
The protein kinase C (PKC) family of serine/threonine kinases consists of ten different isoforms grouped into three subfamilies, denoted classical, novel and atypical PKCs (aPKCs). The aPKCs, PKCι/λ and PKCζ serve important roles during development and in processes subverted in cancer such as cell and tissue polarity, cell proliferation, differentiation and apoptosis. In an effort to identify novel interaction partners for aPKCs, we performed a yeast two-hybrid screen with the regulatory domain of PKCι/λ as bait and identified the Krüppel-like factors family protein TIEG1 as a putative interaction partner for PKCι/λ. We confirmed the interaction of both aPKCs with TIEG1 in vitro and in cells, and found that both aPKCs phosphorylate the DNA-binding domain of TIEG1 on two critical residues. Interestingly, the aPKC-mediated phosphorylation of TIEG1 affected its DNA-binding activity, subnuclear localization and transactivation potential.
Collapse
Affiliation(s)
- Endalkachew A. Alemu
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsö, 9037 Tromsö, Norway
| | - Eva Sjøttem
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsö, 9037 Tromsö, Norway
| | - Heidi Outzen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsö, 9037 Tromsö, Norway
| | - Kenneth B. Larsen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsö, 9037 Tromsö, Norway
| | - Turid Holm
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsö, 9037 Tromsö, Norway
| | - Geir Bjørkøy
- University College of Sør-Trøndelag, 7006 Trondheim, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsö, 9037 Tromsö, Norway
| |
Collapse
|
50
|
Jiang L, Chen Y, Chan CY, Lu G, Wang H, Li JC, Kung HF. Dynamic transcriptional changes of TIEG1 and TIEG2 during mouse tissue development. Anat Rec (Hoboken) 2010; 293:858-864. [PMID: 20201061 DOI: 10.1002/ar.21108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
TGF-beta-inducible early-response gene (TIEG) is a family of primary response genes induced by TGF-beta, which are well recognized in regulating cellular proliferation and apoptosis. However, their expression profile has never been investigated during embryogenesis in different organs. In this study, we aimed to investigate the transcriptional level of both TIEG1 and TIEG2 during development in various mice organs, including the brain cortex, cerebellum and stem, brain striatum, muscle, heart, liver, kidney, and lung. Quantitative real-time PCR was used to profile the change of transcriptional level of the two TIEG members in the mice tissues at six developmental stages. Taken together, the expression of TIEG1 and TIEG2 was specific in different organs yet varied with different developmental time points. Their dynamic changes were moderately consistent in most organs including the brain cortex, striatum, liver, kidney, and lung. However, their mRNA expression in both the heart and muscle was significantly different at all developmental stages, which might propose a compensation of functions in the TIEG family. Nevertheless, our data indicate that both the TIEG genes are essential in regulating the normal organ development and functioning in murine model, as their expressions were ubiquitous and tissue specific at various developmental stages.
Collapse
Affiliation(s)
- Lei Jiang
- Institute of Cell Biology, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|