1
|
Liu Y, Cheng W, Zhao Y, Gao L, Chang Y, Tong Z, Li H, Jing J. Cyclic Mechanical Strain Regulates Osteoblastic Differentiation of Mesenchymal Stem Cells on TiO 2 Nanotubes Through GCN5 and Wnt/β-Catenin. Front Bioeng Biotechnol 2021; 9:735949. [PMID: 34869255 PMCID: PMC8634263 DOI: 10.3389/fbioe.2021.735949] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 09/30/2021] [Indexed: 02/03/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) play a critical role in bone formation and are extremely sensitive to external mechanical stimuli. Mechanical signals can regulate the biological behavior of cells on the surface of titanium-related prostheses and inducing osteogenic differentiation of BMSCs, which provides the integration of host bone and prosthesis benefits. But the mechanism is still unclear. In this study, BMSCs planted on the surface of TiO2 nanotubes were subjected to cyclic mechanical stress, and the related mechanisms were explored. The results of alkaline phosphatase staining, real-time PCR, and Western blot showed that cyclic mechanical stress can regulate the expression level of osteogenic differentiation markers in BMSCs on the surface of TiO2 nanotubes through Wnt/β-catenin. As an important member of the histone acetyltransferase family, GCN5 exerted regulatory effects on receiving mechanical signals. The results of the ChIP assay indicated that GCN5 could activate the Wnt promoter region. Hence, we concluded that the osteogenic differentiation ability of BMSCs on the surface of TiO2 nanotubes was enhanced under the stimulation of cyclic mechanical stress, and GCN5 mediated this process through Wnt/β-catenin.
Collapse
Affiliation(s)
- Yanchang Liu
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Wendan Cheng
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yao Zhao
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Liang Gao
- Sino Euro Orthopaedics Network, Berlin, Germany
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhicheng Tong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Juehua Jing
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
H3K18Ac as a Marker of Cancer Progression and Potential Target of Anti-Cancer Therapy. Cells 2019; 8:cells8050485. [PMID: 31121824 PMCID: PMC6562857 DOI: 10.3390/cells8050485] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
Acetylation and deacetylation are posttranslational modifications (PTMs) which affect the regulation of chromatin structure and its remodeling. Acetylation of histone 3 at lysine placed on position 18 (H3K18Ac) plays an important role in driving progression of many types of cancer, including breast, colon, lung, hepatocellular, pancreatic, prostate, and thyroid cancer. The aim of this review is to analyze and discuss the newest findings regarding the role of H3K18Ac and acetylation of other histones in carcinogenesis. We summarize the level of H3K18Ac in different cancer cell lines and analyze its association with patients’ outcomes, including overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS). Finally, we describe future perspectives of cancer therapeutic strategies based on H3K18 modifications.
Collapse
|
3
|
Mota MSV, Jackson WP, Bailey SK, Vayalil P, Landar A, Rostas JW, Mulekar MS, Samant RS, Shevde LA. Deficiency of tumor suppressor Merlin facilitates metabolic adaptation by co-operative engagement of SMAD-Hippo signaling in breast cancer. Carcinogenesis 2018; 39:1165-1175. [PMID: 29893810 PMCID: PMC6148973 DOI: 10.1093/carcin/bgy078] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/15/2018] [Accepted: 06/08/2018] [Indexed: 12/17/2022] Open
Abstract
The NF2 gene encodes the tumor and metastasis suppressor protein Merlin. Merlin exerts its tumor suppressive role by inhibiting proliferation and inducing contact-growth inhibition and apoptosis. In the current investigation, we determined that loss of Merlin in breast cancer tissues is concordant with the loss of the inhibitory SMAD, SMAD7, of the TGF-β pathway. This was reflected as dysregulated activation of TGF-β signaling that co-operatively engaged with effectors of the Hippo pathway (YAP/TAZ/TEAD). As a consequence, the loss of Merlin in breast cancer resulted in a significant metabolic and bioenergetic adaptation of cells characterized by increased aerobic glycolysis and decreased oxygen consumption. Mechanistically, we determined that the co-operative activity of the Hippo and TGF-β transcription effectors caused upregulation of the long non-coding RNA Urothelial Cancer-Associated 1 (UCA1) that disengaged Merlin's check on STAT3 activity. The consequent upregulation of Hexokinase 2 (HK2) enabled a metabolic shift towards aerobic glycolysis. In fact, Merlin deficiency engendered cellular dependence on this metabolic adaptation, endorsing a critical role for Merlin in regulating cellular metabolism. This is the first report of Merlin functioning as a molecular restraint on cellular metabolism. Thus, breast cancer patients whose tumors demonstrate concordant loss of Merlin and SMAD7 may benefit from an approach of incorporating STAT3 inhibitors.
Collapse
Affiliation(s)
- Mateus S V Mota
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - William P Jackson
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Sarah K Bailey
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Praveen Vayalil
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Aimee Landar
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Jack W Rostas
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Madhuri S Mulekar
- Department of Mathematics and Statistics, University of South Alabama, Mobile, AL, USA
| | - Rajeev S Samant
- Department of Pathology, University of Louisville, Louisville, KY, USA
- UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Louisville, Louisville, KY, USA
- UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
4
|
Up-regulation of HO-1 promotes resistance of B-cell acute lymphocytic leukemia cells to HDAC4/5 inhibitor LMK-235 via the Smad7 pathway. Life Sci 2018; 207:386-394. [PMID: 29886060 DOI: 10.1016/j.lfs.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/22/2018] [Accepted: 06/04/2018] [Indexed: 02/03/2023]
Abstract
PURPOSE HDAC4/5 and Smad7 are potential therapeutic targets for the onset and progression of B-cell acute lymphocytic leukemia (B-ALL) and indices for clinical prognosis. In contrast, HO-1 (heat shock protein 32) plays a key role in protecting tumor cells from apoptosis. METHODS HDAC4/5, HO-1 and Smad7 expressions in 34 newly diagnosed B-ALL cases were detected by real-time PCR and Western blot. Lentivirus and small interference RNA were used to transfect B-ALL cells. The expression of Smad7 was detected after treatment with LMK-235 or Hemin and ZnPP. Apoptosis and proliferation were evaluated by flow cytometry, CCK-8 assay and Western blot. RESULTS HDAC4/5 was overexpressed in B-ALL patients with high HO-1 levels. Increasing the concentration of HDAC4/5 inhibitor LMK-235 induced the decrease of Smad7 and HO-1 expressions and the apoptosis of B-ALL cells by suppressing the phosphorylation of AKT (Protein kinase B). Up-regulating HO-1 alleviated the decrease of Smad7 expression and enhanced B-ALL resistance to LMK-235 by activating p-AKT which reduced the apoptosis of B-ALL cells and influenced the survival of leukemia patients. Silencing Smad7 also augmented the apoptosis rate of B-ALL cells by suppressing p-AKT. CONCLUSION HO-1 played a key role in protecting tumor cells from apoptosis, and HDAC4/5 were related with the apoptosis of B-ALL cells. LMK-235 may be able to improve the poor survival of leukemia patients.
Collapse
|
5
|
Hepatic Smad7 overexpression causes severe iron overload in mice. Blood 2017; 131:581-585. [PMID: 29237592 DOI: 10.1182/blood-2017-07-796797] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 12/02/2017] [Indexed: 02/07/2023] Open
|
6
|
Sferra R, Pompili S, Festuccia C, Marampon F, Gravina GL, Ventura L, Di Cesare E, Cicchinelli S, Gaudio E, Vetuschi A. The possible prognostic role of histone deacetylase and transforming growth factor β/Smad signaling in high grade gliomas treated by radio-chemotherapy: a preliminary immunohistochemical study. Eur J Histochem 2017; 61:2732. [PMID: 28735518 PMCID: PMC5439439 DOI: 10.4081/ejh.2017.2732] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive tumor of the central nervous system. Unfortunately, patients affected by this disease have a very poor prognosis, due to high level of invasiveness and resistance to standard therapies. Although the molecular profile of GBM has been extensively investigated, the events responsible for its pathogenesis and progression remain largely unknown. Histone Deacetylases (HDAC) dependent epigenetic modifications and transforming growth factor (TGF)-β/Smad pathway seem to play an important role in GBM tumorigenesis, resistance to common therapies and poor clinical outcome. The aim of this study was to evaluate the involvement and the possible interaction between these two molecular cascades in the pathogenesis and prognosis of GBM. Immunohistochemistry (IHC) was performed on microdissected GBM samples, collected from 14 patients (6 men and 8 women) ranging in age from 43 to 74 years. The patients were previously divided, on the basis of their overall survival (OS), into two groups: short and long OS. Patients with poor prognosis showed hyperexpression of HDAC4 and HDAC6, an activation of the TGF-β/Smad pathway, with high levels of IL-13, SMAD2, PDGF and MMP3 expression, compared to the long survivors. The short OS group exhibits a decrease in SMAD7 expression and also low levels of p21 immunostaining, which represents a common target of the two pathways. The IHC data was confirmed by quantitative analysis and Immunoblotting. Our preliminary results suggest that both HDAC4 and HDAC6 together with the TGF-β/Smad pathway may be involved in progression of GBM and this cross talking could be a useful prognostic marker in this deadly disease.
Collapse
Affiliation(s)
- Roberta Sferra
- University of L'Aquila, Department of Biotechnological and Applied Clinical Sciences.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Zhou W, Chen X, He K, Xiao J, Duan X, Huang R, Xia Z, He J, Zhang J, Xiang G. Histone deacetylase inhibitor screening identifies HC toxin as the most effective in intrahepatic cholangiocarcinoma cells. Oncol Rep 2016; 35:2535-42. [PMID: 26935789 PMCID: PMC4811396 DOI: 10.3892/or.2016.4636] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 12/17/2015] [Indexed: 01/04/2023] Open
Abstract
Histone deacetylases (HDACs) are highly expressed in intrahepatic cholangiocarcinoma (ICC) and are associated with poor prognosis of these patients. The aim of the present study was to explore the inhibitory effects of HDAC inhibitors on ICC cells and identify effective and sensitive drugs for ICC. Effects of 34 HDAC inhibitors were screened through two rounds of cell viability assays, and HC toxin, a cyclic tetrapeptide first isolated from the secondary metabolite of Helminthosporium carbonum, exhibited an antitumor activity superior to that of the other HDAC inhibitors and gemcitabine. The mechanisms involved in the inhibitory effects of HC toxin on CCLP-1 cells were investigated by cell counting, colony formation assay, cell morphological observation, real-time PCR, western blotting and flow cytometry. It was demonstrated that HC toxin inhibited the cell proliferation and clone formation ability of the CCLP-1 cells. HC toxin increased the acetyl-histone H4 level and this was associated with the inhibitory effect of HC toxin on the CCLP-1 cells. We also found that HC toxin reduced the level of HDAC1 protein in a post-transcriptional manner. Morphological observation showed multiple morphological changes and indicated the possibility of cell differentiation owing to HC toxin. With increasing concentration of HC toxin, the cell cycle was gradually arrested at the G0/G1 stage and the percentage of apoptotic cells increased which was not mainly through the caspase-3-dependent ways. These results indicated that HC toxin was the most effective among the various HDAC inhibitors with multiple functions in the suppression of ICC in vitro. Thus, HC may be a potential chemotherapeutic for ICC.
Collapse
Affiliation(s)
- Wenjie Zhou
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Xiaoxun Chen
- Department of Gastrointestinal Surgery, The Guigang City People's Hospital, Guigang, Guangxi 537100, P.R. China
| | - Ke He
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Jinfeng Xiao
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Xiaopeng Duan
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Rui Huang
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Zhenglin Xia
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Jingliang He
- Graduate School, Guangdong Medical College, Zhanjiang, Guangdong 524023, P.R. China
| | - Jinqian Zhang
- Department of Laboratory Medicine, The Second People's Hospital of Guangdong Province, Guangzhou, Guangdong 510317, P.R. China
| | - Guoan Xiang
- Department of General Surgery, The Second People's Hospital of Guangdong Province, The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| |
Collapse
|
8
|
Hua C, Wang Z, Zhang J, Peng X, Hou X, Yang Y, Li K, Tang Z. SMAD7, an antagonist of TGF-beta signaling, is a candidate of prenatal skeletal muscle development and weaning weight in pigs. Mol Biol Rep 2016; 43:241-51. [DOI: 10.1007/s11033-016-3960-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/17/2016] [Indexed: 12/22/2022]
|
9
|
Li S, Liu X, Chen X, Zhang L, Wang X. Histone deacetylase 6 promotes growth of glioblastoma through inhibition of SMAD2 signaling. Tumour Biol 2015; 36:9661-5. [DOI: 10.1007/s13277-015-3747-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/29/2014] [Indexed: 11/28/2022] Open
|
10
|
Luo L, Li N, Lv N, Huang D. SMAD7: a timer of tumor progression targeting TGF-β signaling. Tumour Biol 2014; 35:8379-85. [PMID: 24935472 DOI: 10.1007/s13277-014-2203-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/06/2014] [Indexed: 01/02/2023] Open
Abstract
In the context of cancer, transforming growth factor β (TGF-β) is a cell growth suppressor; however, it is also a critical inducer of invasion and metastasis. SMAD is the important mediator of TGF-β signaling pathway, which includes receptor-regulated SMADs (R-SMADs), common-mediator SMADs (co-SMADs), and inhibitory SMADs (I-SMADs). I-SMADs block the activation of R-SMADs and co-SMADs and thus play important roles especially in the SMAD-dependent signaling. SMAD7 belongs to the I-SMADs. As an inhibitor of TGF-β signaling, SMAD7 is overexpressed in numerous cancer types and its abundance is positively correlated to the malignancy. Emerging evidence has revealed the switch-in-role of SMAD7 in cancer, from a TGF-β inhibiting protein at the early stages that facilitates proliferation to an enhancer of invasion at the late stages. This role change may be accompanied or elicited by the tumor microenvironment and/or somatic mutation. Hence, current knowledge suggests a tumor-favorable timer nature of SMAD7 in cancer progression. In this review, we summarized the advances and recent findings of SMAD7 and TGF-β signaling in cancer, followed by specific discussion on the possible factors that account for the functional changes of SMAD7.
Collapse
Affiliation(s)
- Lingyu Luo
- Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, 17th Yongwaizen St., Nanchang, Jiangxi, 330006, People's Republic of China
| | | | | | | |
Collapse
|
11
|
Yin Yang 1 (YY1) synergizes with Smad7 to inhibit TGF-β signaling in the nucleus. SCIENCE CHINA-LIFE SCIENCES 2013; 57:128-36. [PMID: 24369345 DOI: 10.1007/s11427-013-4581-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 11/19/2013] [Indexed: 10/25/2022]
Abstract
As a prototype of the TGF-β superfamily cytokines, TGF-β is well known for its diverse roles in embryogenesis and adult tissue homeostasis. TGF-β evokes cellular responses by signaling mainly through cell membrane receptors and transcription factor R-Smads and Co-Smad (Smad4), while an inhibitory Smad, Smad7, acts as a critical negative regulator of TGF-β signaling. Smad7 antagonizes TGF-β signaling by regulating the stability or activity of the receptors or blocking the DNA binding of the functional R-Smad-Smad4 complex in the nucleus. However, the function of Smad7 in the nucleus is not fully understood. Yin Yang 1 (YY1) is a ubiquitously expressed transcription factor with multiple functions. It has been reported that YY1 can inhibit Smad-dependent transcriptional responses and TGF-β/BMP-induced cell differentiation independently of its DNA binding ability. In this study, we found that Smad7 interacts with YY1 and the interaction is attenuated by TGF-β signaling. Reporter assays and target gene expression analyses revealed that Smad7 and YY1 act in concert to inhibit TGF-β-induced transcription in the nucleus. Furthermore, Smad7 could enhance the interaction of YY1 with the histone deacetylase HDAC1. Consistently, YY1 and HDAC1 augmented the transcription repression activity of Smad7 in Gal4-luciferase reporter analysis. Therefore, our findings define a novel mechanism of Smad7 and YY1 to antagonize TGF-β signaling.
Collapse
|
12
|
Wang M, Saha J, Cucinotta FA. Smad7 foci are present in micronuclei induced by heavy particle radiation. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2013; 756:108-14. [DOI: 10.1016/j.mrgentox.2013.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 04/23/2013] [Indexed: 12/13/2022]
|
13
|
High SKIP expression is correlated with poor prognosis and cell proliferation of hepatocellular carcinoma. Med Oncol 2013; 30:537. [PMID: 23696020 DOI: 10.1007/s12032-013-0537-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 03/05/2013] [Indexed: 02/08/2023]
Abstract
Ski-interacting protein (SKIP) is a transcriptional cofactor distinct from other cofactors and involved in the regulation of many cancer-related proteins. This study investigated the expression of SKIP and its potential clinical and biological significances in hepatocellular carcinoma (HCC). Immunohistochemistry and Western blot were performed to detect the expression of SKIP in clinical HCC samples and adjacent noncancerous tissues. In addition, expression of SKIP was correlated with clinicopathological variables, and univariate and multivariate survival analyses were performed to determine the prognostic significance. Moreover, the biological significance of the aberrant expression of SKIP was investigated in vitro. High SKIP expression was detected in clinical HCC samples compared with adjacent noncancerous tissues. Expression of SKIP correlated directly with the histological grades of HCC and high expression of SKIP was associated with a poor prognosis. SKIP depletion by small interfering RNA inhibited cell proliferation and blocked S phase entry in HepG2 cells. Owing to overexpression of SKIP in HCC tissues and its important role in predicting poor prognosis and the development of HCC, SKIP could be a potential prognostic marker and therapeutic target of HCC.
Collapse
|