1
|
Han H, zhang C, Shi W, Wang J, Zhao W, Du Y, Zhao Z, Wang Y, Lin M, Qin L, Zhao X, Yin Q, Liu Y, Wang Z, Zhang J, You X, Zhou G, Wu H, Ye J, He X, Tian W, Yu H, Yuan Y, Wang Q. NSUN5 Facilitates Hepatocellular Carcinoma Progression by Increasing SMAD3 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404083. [PMID: 39531371 PMCID: PMC11727281 DOI: 10.1002/advs.202404083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/06/2024] [Indexed: 11/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by frequent intrahepatic and distant metastases, resulting in a poor prognosis for patients. Epithelial-mesenchymal transition (EMT) plays a pivotal role in this process. However, the expression of NOP2/Sun RNA methyltransferase 5 (NSUN5) in HCC and its role in mediating EMT remain poorly understood. In this study, clinicopathological analyses are conducted across multiple independent HCC cohorts and induced tumor formation in Nsun5-knockout mice. The findings reveal an upregulation of NSUN5 expression in tumor tissues; conversely, the absence of Nsun5 hinders the malignant progression of HCC, indicating that NSUN5 may serve as a significant oncogene in HCC. Furthermore, elevated levels of NSUN5 enhance EMT processes within HCC cells. NSUN5-knockout cells exhibit reduced invasion and migration capabilities under both in vivo and in vitro conditions, while overexpression of NSUN5 yields opposing effects. Mechanistically, high levels of NSUN5 promote the enrichment of trimethylated histone H3 at lysine 4 (H3K4me3) at the promoter region of SMAD3 through recruitment of the WDR5, thereby facilitating HCC metastasis via SMAD3-mediated EMT pathways. Collectively, this study identifies NSUN5 as a novel driver of metastasis in HCC and provides a theoretical foundation for potential therapeutic strategies against this malignancy.
Collapse
Affiliation(s)
- Hexu Han
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Chengcheng zhang
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Wenbo Shi
- Oncology majorRuijin‐Hainan HospitalShanghai Jiao Tong University School of MedicineHainan200032China
| | - Jiawei Wang
- Department of Hepatobiliary SurgeryThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
- Department of General SurgeryFirst Affiliated Hospital of Suzhou UniversitySuzhou215000P. R. China
| | - Wenhui Zhao
- Department of Basic MedicineJiangsu College of NursingHuai'anJiangsu223001P. R. China
| | - Yanping Du
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Zhibin Zhao
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Yifan Wang
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Maosong Lin
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Lei Qin
- Department of Hepatobiliary SurgeryThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
- Department of General SurgeryFirst Affiliated Hospital of Suzhou UniversitySuzhou215000P. R. China
| | - Xiaoxue Zhao
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Qianqian Yin
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Yiyi Liu
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Zhongqi Wang
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong UniversityNantong UniversityJiangsu226001P. R. China
| | - Xiaomin You
- Department of Gastroenterology, Affiliated Hospital of Nantong UniversityNantong UniversityJiangsu226001P. R. China
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong UniversityNantong UniversityJiangsu226001P. R. China
| | - Honghui Wu
- Reproduction Medicine CentreThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou225300P. R. China
| | - Jun Ye
- Center for Translational Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Xianzhong He
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityInnovative Institute of Tumor Immunity and Medicine (ITIM)Anhui Provincial Innovation Institute for Pharmaceutical Basic ResearchAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230000P. R. China
| | - Weizhong Tian
- Department of RadiologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Hong Yu
- Department of PathologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Yin Yuan
- Department of Hepatobiliary SurgeryThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Qiang Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityInnovative Institute of Tumor Immunity and Medicine (ITIM)Anhui Provincial Innovation Institute for Pharmaceutical Basic ResearchAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230000P. R. China
| |
Collapse
|
2
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
3
|
Radak M, Ghamari N, Fallahi H. Identification of common factors among fibrosarcoma, rhabdomyosarcoma, and osteosarcoma by network analysis. Biosystems 2024; 235:105093. [PMID: 38052344 DOI: 10.1016/j.biosystems.2023.105093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023]
Abstract
Sarcoma cancers are uncommon malignant tumors, and there are many subgroups, including fibrosarcoma (FS), which mainly affects middle-aged and older adults in deep soft tissues. Rhabdomyosarcoma (RMS), on the other hand, is the most common soft-tissue sarcoma in children and is located in the head and neck area. Osteosarcomas (OS) is the predominant form of primary bone cancer among young adults, primarily resulting from sporadically random mutations. This frequently results in the dissemination of cancer cells to the lungs, commonly known as metastasis. Mesodermal cells are the origin of sarcoma cancers. In this study, a rather radical approach has been applied. Instead of comparing homogenous cancer types, we focus on three main subtypes of sarcoma: fibrosarcoma, rhabdomyosarcoma, and osteosarcoma, and compare their gene expression with normal cell groups to identify the differentially expressed genes (DEGs). Next, by applying protein-protein interaction (PPI) network analysis, we determine the hub genes and crucial factors, such as transcription factors (TFs), affected by these types of cancer. Our findings indicate a modification in a range of pathways associated with cell cycle, extracellular matrix, and DNA repair in these three malignancies. Results showed that fibrosarcoma (FS), rhabdomyosarcoma (RMS), and osteosarcoma (OS) had 653, 1270, and 2823 differentially expressed genes (DEGs), respectively. Interestingly, there were 24 DEGs common to all three types. Network analysis showed that the fibrosarcoma network had two sub-networks identified in FS that contributed to the catabolic process of collagen via the G-protein coupled receptor signaling pathway. The rhabdomyosarcoma network included nine sub-networks associated with cell division, extracellular matrix organization, mRNA splicing via spliceosome, and others. The osteosarcoma network has 13 sub-networks, including mRNA splicing, sister chromatid cohesion, DNA repair, etc. In conclusion, the common DEGs identified in this study have been shown to play significant and multiple roles in various other cancers based on the literature review, indicating their significance.
Collapse
Affiliation(s)
- Mehran Radak
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Iran.
| | - Nakisa Ghamari
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Iran.
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Iran.
| |
Collapse
|
4
|
Kim JY, Hong N, Park S, Ham SW, Kim EJ, Kim SO, Jang J, Kim Y, Kim JK, Kim SC, Park JW, Kim H. Jagged1 intracellular domain/SMAD3 complex transcriptionally regulates TWIST1 to drive glioma invasion. Cell Death Dis 2023; 14:822. [PMID: 38092725 PMCID: PMC10719344 DOI: 10.1038/s41419-023-06356-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Jagged1 (JAG1) is a Notch ligand that correlates with tumor progression. Not limited to its function as a ligand, JAG1 can be cleaved, and its intracellular domain translocates to the nucleus, where it functions as a transcriptional cofactor. Previously, we showed that JAG1 intracellular domain (JICD1) forms a protein complex with DDX17/SMAD3/TGIF2. However, the molecular mechanisms underlying JICD1-mediated tumor aggressiveness remains unclear. Here, we demonstrate that JICD1 enhances the invasive phenotypes of glioblastoma cells by transcriptionally activating epithelial-to-mesenchymal transition (EMT)-related genes, especially TWIST1. The inhibition of TWIST1 reduced JICD1-driven tumor aggressiveness. Although SMAD3 is an important component of transforming growth factor (TGF)-β signaling, the JICD1/SMAD3 transcriptional complex was shown to govern brain tumor invasion independent of TGF-β signaling. Moreover, JICD1-TWIST1-MMP2 and MMP9 axes were significantly correlated with clinical outcome of glioblastoma patients. Collectively, we identified the JICD1/SMAD3-TWIST1 axis as a novel inducer of invasive phenotypes in cancer cells.
Collapse
Affiliation(s)
- Jung Yun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Nayoung Hong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sehyeon Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Seok Won Ham
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Eun-Jung Kim
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Sung-Ok Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Junseok Jang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoonji Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Jun-Kyum Kim
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Sung-Chan Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Jong-Whi Park
- Department of Life Sciences, Gachon University, Incheon, 21999, Republic of Korea.
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
5
|
Matsumoto Y, Fukui T, Horitani S, Tanimura Y, Suzuki R, Tomiyama T, Honzawa Y, Tahara T, Okazaki K, Naganuma M. A Short-Term Model of Colitis-Associated Colorectal Cancer That Suggests Initial Tumor Development and the Characteristics of Cancer Stem Cells. Int J Mol Sci 2023; 24:11697. [PMID: 37511456 PMCID: PMC10380789 DOI: 10.3390/ijms241411697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
The mechanisms underlying the transition from colitis-associated inflammation to carcinogenesis and the cell origin of cancer formation are still unclear. The azoxymethane (AOM)/dextran sodium sulfate (DSS) mouse model reproduces human colitis-associated colorectal cancer. To elucidate the mechanisms of cancer development and dynamics of the linker threonine-phosphorylated Smad2/3 (pSmad2/3L-Thr)-positive cells, we explored the early stages of colitis-associated colorectal cancer in AOM/DSS mice. The AOM/DSS mice were sacrificed at 4 to 6 weeks following AOM administration. To analyze the initial lesions, immunofluorescence staining for the following markers was performed: β-catenin, Ki67, CDK4, Sox9, Bmi1, cyclin D1, and pSmad2/3L-Thr. Micro-neoplastic lesions were flat and unrecognizable, and the uni-cryptal ones were either open to the surfaces or hidden within the mucosae. These neoplastic cells overexpressed β-catenin, Sox9, Ki67, and Cyclin D1 and had large basophilic nuclei in the immature atypical cells. In both the lesions, pSmad2/3L-Thr-positive cells were scattered and showed immunohistochemical co-localization with β-catenin, CDK4, and Bmi1 but never with Ki67. More β-catenin-positive neoplastic cells of both lesions were detected at the top compared to the base or center of the mucosae. We confirmed initial lesions in the colitis-associated colorectal cancer model mice and observed results that suggest that pSmad2/3L-Thr is a biomarker for tissue stem cells and cancer stem cells.
Collapse
Affiliation(s)
- Yasushi Matsumoto
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Toshiro Fukui
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Shunsuke Horitani
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yuji Tanimura
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Ryo Suzuki
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Takashi Tomiyama
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yusuke Honzawa
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Tomomitsu Tahara
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Kazuichi Okazaki
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| | - Makoto Naganuma
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Japan
| |
Collapse
|
6
|
Wang C, Zhang Y, Zhao D, Huo Y, Xie J, Zhang X, Luo H, Xu H, Zhang YW. Phenazopyridine promotes RPS23RG1/Rps23rg1 transcription and ameliorates Alzheimer-associated phenotypes in mice. Neuropsychopharmacology 2022; 47:2042-2050. [PMID: 35821069 PMCID: PMC9556769 DOI: 10.1038/s41386-022-01373-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 06/05/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with no effective treatment options. A complete elucidation of its underlying molecular mechanisms, including the transcription regulation of genes critically involved in AD, may shed light on new therapeutic development. RPS23RG1 is a newly identified AD-associated gene, whose expression is decreased in AD and restoration can attenuate AD-like phenotypes in animal models. However, the transcription regulation of RPS23RG1 remains unknown. In this study, we explored the promoter of RPS23RG1 and identified its transcription initiation site (TSS) at 1525 bp upstream of the ATG translation start codon. Progressive deletion analysis determined the presence of a negative regulatory region and a positive regulatory region within nucleotide positions +1127 to +1187 and +732 to +1127 relative to the TSS (+1), respectively. We conducted a reporter system to screen for compounds that increase RPS23RG1 expression through antagonizing its negative regulatory elements and identified phenazopyridine. Importantly, we demonstrated that phenazopyridine not only promoted RPS23RG1/Rps23rg1 expression, but also reduced AD-like pathologies and cognitive impairments in the APP/PS1 AD model mice. We also determined a critical negative regulatory domain of RPS23RG1 within nucleotide positions +1177 to +1187 and found that the transcription factor SMAD3 bound to this domain. Inhibition of SMAD3 promoted RPS23RG1 expression. Moreover, phenazopyridine reduced SMAD3 binding to the RPS23RG1 promoter without affecting SMAD3 phosphorylation and nuclear localization. Taken together, our results determine the transcription regulation mechanism of RPS23RG1 and show that phenazopyridine has potential for AD treatment through regulating RPS23RG1 transcription.
Collapse
Affiliation(s)
- Chong Wang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China.
| | - Yuan Zhang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Dongdong Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Yuanhui Huo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Jieru Xie
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Xian Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Hong Luo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Huaxi Xu
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, PR China.
| |
Collapse
|
7
|
Chen S, Shu G, Wang G, Ye J, Xu J, Huang C, Yang S. HOXA1 promotes proliferation and metastasis of bladder cancer by enhancing SMAD3 transcription. Pathol Res Pract 2022; 239:154141. [DOI: 10.1016/j.prp.2022.154141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/22/2022] [Accepted: 09/25/2022] [Indexed: 11/28/2022]
|
8
|
Li LX, Agborbesong E, Zhang L, Zhang X, Zhou JX, Li X. Crosstalk between lysine methyltransferase Smyd2 and TGF-β-Smad3 signaling promotes renal fibrosis in autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2022; 323:F227-F242. [PMID: 35759739 PMCID: PMC9359663 DOI: 10.1152/ajprenal.00452.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited genetic disorder, which is caused by mutations of PKD1 or PKD2 gene and is characterized by renal fluid-filled cyst formation and interstitial fibrosis. PKD1 gene mutation results in the upregulation of SET and MYND domain-containing lysine methyltransferase 2 (SMYD2) in Pkd1 mutant mouse and ADPKD patient kidneys. However, the role and mechanism of Smyd2 in the regulation of renal fibrosis in ADPKD remains elusive. In this study, we show that: 1) the expression of Smyd2 can be regulated by TGF-β-Smad3 in normal rat kidney 49F (NRK-49F) cells and mouse fibroblast NIH3T3 cells; 2) knockdown of Smyd2 and inhibition of Smyd2 with its specific inhibitor, AZ505, decreases TGF-β-induced expression of α-smooth muscle actin (α-SMA), fibronectin, collagens 1 and 3 and plasminogen activator inhibitor-1( PAI1) in NRK-49F cells; 3) Smyd2 regulates the transcription of fibrotic marker genes through binding on the promoters of those genes or through methylating histone H3 to indirectly regulate the expression of those genes; and 4) knockout and inhibition of Smyd2 significantly decreases renal fibrosis in Pkd1 knockout mice, supporting that targeting Smyd2 can not only delay cyst growth but also attenuate renal fibrosis in ADPKD. This study identifies a crosstalk between TGF-β signaling and Smyd2 in the regulation of fibrotic gene transcription and the activation of fibroblasts in cystic kidneys, suggesting that targeting Smyd2 with AZ505 is a potential therapeutic strategy for ADPKD treatment.
Collapse
Affiliation(s)
- Linda Xiaoyan Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Lu Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Xiaoqin Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Julie Xia Zhou
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
9
|
Chaperone-mediated Autophagy Regulates Cell Growth by Targeting SMAD3 in Glioma. Neurosci Bull 2022; 38:637-651. [PMID: 35267139 DOI: 10.1007/s12264-022-00818-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/10/2021] [Indexed: 10/18/2022] Open
Abstract
Previous studies suggest that the reduction of SMAD3 (mothers against decapentaplegic homolog 3) has a great impact on tumor development, but its exact pathological function remains unclear. In this study, we found that the protein level of SMAD3 was greatly reduced in human-grade IV glioblastoma tissues, in which LAMP2A (lysosome-associated membrane protein type 2A) was significantly up-regulated. LAMP2A is a key rate-limiting protein of chaperone-mediated autophagy (CMA), a lysosome pathway of protein degradation that is activated in glioma. We carefully analyzed the amino-acid sequence of SMAD3 and found that it contained a pentapeptide motif biochemically related to KFERQ, which has been proposed to be a targeting sequence for CMA. In vitro, we confirmed that SMAD3 was degraded in either serum-free or KFERQ motif deleted condition, which was regulated by LAMP2A and interacted with HSC70 (heat shock cognate 71 kDa protein). Using isolated lysosomes, amino-acid residues 75 and 128 of SMAD3 were found to be of importance for this process, which affected the CMA pathway in which SMAD3 was involved. Similarly, down-regulating SMAD3 or up-regulating LAMP2A in cultured glioma cells enhanced their proliferation and invasion. Taken together, these results suggest that excessive activation of CMA regulates glioma cell growth by promoting the degradation of SMAD3. Therefore, targeting the SMAD3-LAMP2A-mediated CMA-lysosome pathway may be a promising approach in anti-cancer therapy.
Collapse
|
10
|
Decker JT, Ma JA, Shea LD, Jeruss JS. Implications of TGFβ Signaling and CDK Inhibition for the Treatment of Breast Cancer. Cancers (Basel) 2021; 13:5343. [PMID: 34771508 PMCID: PMC8582459 DOI: 10.3390/cancers13215343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 01/01/2023] Open
Abstract
TGFβ signaling enacts tumor-suppressive functions in normal cells through promotion of several cell regulatory actions including cell-cycle control and apoptosis. Canonical TGFβ signaling proceeds through phosphorylation of the transcription factor, SMAD3, at the C-terminus of the protein. During oncogenic progression, this tumor suppressant phosphorylation of SMAD3 can be inhibited. Overexpression of cyclins D and E, and subsequent hyperactivation of cyclin-dependent kinases 2/4 (CDKs), are often observed in breast cancer, and have been associated with poor prognosis. The noncanonical phosphorylation of SMAD3 by CDKs 2 and 4 leads to the inhibition of tumor-suppressive function of SMAD3. As a result, CDK overactivation drives oncogenic progression, and can be targeted to improve clinical outcomes. This review focuses on breast cancer, and highlights advances in the understanding of CDK-mediated noncanonical SMAD3 phosphorylation. Specifically, the role of aberrant TGFβ signaling in oncogenic progression and treatment response will be examined to illustrate the potential for therapeutic discovery in the context of cyclins/CDKs and SMAD3.
Collapse
Affiliation(s)
- Joseph T. Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (J.T.D.); (J.A.M.); (L.D.S.)
| | - Jeffrey A. Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (J.T.D.); (J.A.M.); (L.D.S.)
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (J.T.D.); (J.A.M.); (L.D.S.)
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109-5932, USA
| | - Jacqueline S. Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (J.T.D.); (J.A.M.); (L.D.S.)
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109-5932, USA
| |
Collapse
|
11
|
TGF-β/activin signaling promotes CDK7 inhibitor resistance in triple-negative breast cancer cells through upregulation of multidrug transporters. J Biol Chem 2021; 297:101162. [PMID: 34481843 PMCID: PMC8498470 DOI: 10.1016/j.jbc.2021.101162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Cyclin-dependent kinase 7 (CDK7) is a master regulatory kinase that drives cell cycle progression and stimulates expression of oncogenes in a myriad of cancers. Inhibitors of CDK7 (CDK7i) are currently in clinical trials; however, as with many cancer therapies, patients will most likely experience recurrent disease due to acquired resistance. Identifying targets underlying CDK7i resistance will facilitate prospective development of new therapies that can circumvent such resistance. Here we utilized triple-negative breast cancer as a model to discern mechanisms of resistance as it has been previously shown to be highly responsive to CDK7 inhibitors. After generating cell lines with acquired resistance, high-throughput RNA sequencing revealed significant upregulation of genes associated with efflux pumps and transforming growth factor-beta (TGF-β) signaling pathways. Genetic silencing or pharmacological inhibition of ABCG2, an efflux pump associated with multidrug resistance, resensitized resistant cells to CDK7i, indicating a reliance on these transporters. Expression of activin A (INHBA), a member of the TGF-β family of ligands, was also induced, whereas its intrinsic inhibitor, follistatin (FST), was repressed. In resistant cells, increased phosphorylation of SMAD3, a downstream mediator, confirmed an increase in activin signaling, and phosphorylated SMAD3 directly bound the ABCG2 promoter regulatory region. Finally, pharmacological inhibition of TGF-β/activin receptors or genetic silencing of SMAD4, a transcriptional partner of SMAD3, reversed the upregulation of ABCG2 in resistant cells and phenocopied ABCG2 inhibition. This study reveals that inhibiting the TGF-β/Activin-ABCG2 pathway is a potential avenue for preventing or overcoming resistance to CDK7 inhibitors.
Collapse
|
12
|
Thielen N, Neefjes M, Wiegertjes R, van den Akker G, Vitters E, van Beuningen H, Blaney Davidson E, Koenders M, van Lent P, van de Loo F, van Caam A, van der Kraan P. Osteoarthritis-Related Inflammation Blocks TGF-β's Protective Effect on Chondrocyte Hypertrophy via (de)Phosphorylation of the SMAD2/3 Linker Region. Int J Mol Sci 2021; 22:ijms22158124. [PMID: 34360888 PMCID: PMC8347103 DOI: 10.3390/ijms22158124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/13/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by irreversible cartilage damage, inflammation and altered chondrocyte phenotype. Transforming growth factor-β (TGF-β) signaling via SMAD2/3 is crucial for blocking hypertrophy. The post-translational modifications of these SMAD proteins in the linker domain regulate their function and these can be triggered by inflammation through the activation of kinases or phosphatases. Therefore, we investigated if OA-related inflammation affects TGF-β signaling via SMAD2/3 linker-modifications in chondrocytes. We found that both Interleukin (IL)-1β and OA-synovium conditioned medium negated SMAD2/3 transcriptional activity in chondrocytes. This inhibition of TGF-β signaling was enhanced if SMAD3 could not be phosphorylated on Ser213 in the linker region and the inhibition by IL-1β was less if the SMAD3 linker could not be phosphorylated at Ser204. Our study shows evidence that inflammation inhibits SMAD2/3 signaling in chondrocytes via SMAD linker (de)-phosphorylation. The involvement of linker region modifications may represent a new therapeutic target for OA.
Collapse
Affiliation(s)
- Nathalie Thielen
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Margot Neefjes
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Renske Wiegertjes
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Guus van den Akker
- Department of Orthopedic Surgery, Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Elly Vitters
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Henk van Beuningen
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Esmeralda Blaney Davidson
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Marije Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Peter van Lent
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Fons van de Loo
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Arjan van Caam
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Peter van der Kraan
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
- Correspondence:
| |
Collapse
|
13
|
Specific Smad2/3 Linker Phosphorylation Indicates Esophageal Non-neoplastic and Neoplastic Stem-Like Cells and Neoplastic Development. Dig Dis Sci 2021; 66:1862-1874. [PMID: 32705438 DOI: 10.1007/s10620-020-06489-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/11/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND There is little known about stem cells in human non-neoplastic and neoplastic esophageal epithelia. We have demonstrated expression of linker threonine-phosphorylated Smad2/3 (pSmad2/3L-Thr), suggesting presence of stem-like cells in mouse esophageal epithelium, and identified presence of pSmad2/3L-Thr-positive cells that might function as cancer stem cells in mouse model of colorectal carcinoma. AIMS We explore whether pSmad2/3L-Thr can be used as a biomarker for stem cells of human esophageal epithelia and/or neoplasms. METHODS We have used esophageal tissues from inpatients undergoing endoscopic submucosal dissection and performed double immunofluorescent staining of pSmad2/3L-Thr and Ki67, CDK4, p63, Sox2, CK14, p53, ALDH1, CD44 or D2-40 after which the sections were stained with hematoxylin and eosin. RESULTS pSmad2/3L-Thr-positive cells showed immunohistochemical co-localization with CDK4, p63, CD44 and Sox2 in the basal and parabasal layers of non-neoplastic esophageal epithelia. In esophageal neoplasms, they showed immunohistochemical co-localization with p53, CDK4, ALDH1 and CD44. There was a significant increase in the percentage of pSmad2/3L-Thr-positive cells in the p53-positive neoplastic cell population with development of esophageal neoplasia. pSmad2/3L-Thr-positive cells localized to the lower section of low-grade intraepithelial neoplasia and were observed up to the upper section in carcinoma in situ. In invasive squamous cell carcinoma, they were scattered throughout the tumor with disappearance of polarity and were found in intraepithelial primary lesions and sites of submucosal and vessel invasion. CONCLUSIONS We determined significant expression of pSmad2/3L-Thr in human esophageal non-neoplastic and neoplastic epithelia, indicating that these are epithelial stem-like cells and cancer stem cells, respectively, that correlate with developing esophageal neoplasms.
Collapse
|
14
|
Azad AKM, Alyami SA. Discovering novel cancer bio-markers in acquired lapatinib resistance using Bayesian methods. Brief Bioinform 2021; 22:6226686. [PMID: 33857297 DOI: 10.1093/bib/bbab137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/20/2021] [Accepted: 03/23/2021] [Indexed: 11/12/2022] Open
Abstract
Signalling transduction pathways (STPs) are commonly hijacked by many cancers for their growth and malignancy, but demystifying their underlying mechanisms is difficult. Here, we developed methodologies with a fully Bayesian approach in discovering novel driver bio-markers in aberrant STPs given high-throughput gene expression (GE) data. This project, namely 'PathTurbEr' (Pathway Perturbation Driver) uses the GE dataset derived from the lapatinib (an EGFR/HER dual inhibitor) sensitive and resistant samples from breast cancer cell lines (SKBR3). Differential expression analysis revealed 512 differentially expressed genes (DEGs) and their pathway enrichment revealed 13 highly perturbed singalling pathways in lapatinib resistance, including PI3K-AKT, Chemokine, Hippo and TGF-$\beta $ singalling pathways. Next, the aberration in TGF-$\beta $ STP was modelled as a causal Bayesian network (BN) using three MCMC sampling methods, i.e. Neighbourhood sampler (NS) and Hit-and-Run (HAR) sampler that potentially yield robust inference with lower chances of getting stuck at local optima and faster convergence compared to other state-of-art methods. Next, we examined the structural features of the optimal BN as a statistical process that generates the global structure using $p_1$-model, a special class of Exponential Random Graph Models (ERGMs), and MCMC methods for their hyper-parameter sampling. This step enabled key drivers identification that drive the aberration within the perturbed BN structure of STP, and yielded 34, 34 and 23 perturbation driver genes out of 80 constituent genes of three perturbed STP models of TGF-$\beta $ signalling inferred by NS, HAR and MH sampling methods, respectively. Functional-relevance and disease-relevance analyses suggested their significant associations with breast cancer progression/resistance.
Collapse
Affiliation(s)
- A K M Azad
- iThree Institute, Faculty of Science, University of Technology Sydney, Ultimo, 2007, NSW, Australia
| | - Salem A Alyami
- Department of Mathematics & Statistics, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Erkeland SJ, Stavast CJ, Schilperoord-Vermeulen J, Dal Collo G, Van de Werken HJG, Leon LG, Van Hoven-Beijen A, Van Zuijen I, Mueller YM, Bindels EM, De Ridder D, Kappers-Klunne MC, Van Lom K, Van der Velden VHJ, Langerak AW. The miR-200c/141-ZEB2-TGFβ axis is aberrant in human T-cell prolymphocytic leukemia. Haematologica 2021; 107:143-153. [PMID: 33596640 PMCID: PMC8719092 DOI: 10.3324/haematol.2020.263756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 11/29/2022] Open
Abstract
T-cell prolymphocytic leukemia (T-PLL) is mostly characterized by aberrant expansion of small- to medium-sized prolymphocytes with a mature post-thymic phenotype, high aggressiveness of the disease and poor prognosis. However, T-PLL is more heterogeneous with a wide range of clinical, morphological, and molecular features, which occasionally impedes the diagnosis. We hypothesized that T-PLL consists of phenotypic and/or genotypic subgroups that may explain the heterogeneity of the disease. Multi-dimensional immuno-phenotyping and gene expression profiling did not reveal clear T-PLL subgroups, and no clear T-cell receptor a or b CDR3 skewing was observed between different T-PLL cases. We revealed that the expression of microRNA (miRNA) is aberrant and often heterogeneous in T-PLL. We identified 35 miRNA that were aberrantly expressed in T-PLL with miR-200c/141 as the most differentially expressed cluster. High miR- 200c/141 and miR-181a/181b expression was significantly correlated with increased white blood cell counts and poor survival. Furthermore, we found that overexpression of miR-200c/141 correlated with downregulation of their targets ZEB2 and TGFbR3 and aberrant TGFb1- induced phosphorylated SMAD2 (p-SMAD2) and p-SMAD3, indicating that the TGFb pathway is affected in T-PLL. Our results thus highlight the potential role for aberrantly expressed oncogenic miRNA in T-PLL and pave the way for new therapeutic targets in this disease.
Collapse
Affiliation(s)
- Stefan J Erkeland
- Department of Immunology, Erasmus University Medical Center, Rotterdam.
| | | | | | - Giada Dal Collo
- Department of Immunology, Erasmus University Medical Center, Rotterdam
| | - Harmen J G Van de Werken
- Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands; Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam
| | - Leticia G Leon
- Department of Immunology, Erasmus University Medical Center, Rotterdam
| | | | - Iris Van Zuijen
- Department of Immunology, Erasmus University Medical Center, Rotterdam
| | - Yvonne M Mueller
- Department of Immunology, Erasmus University Medical Center, Rotterdam
| | - Eric M Bindels
- Department of Hematology, Erasmus University Medical Center, Rotterdam
| | | | | | - Kirsten Van Lom
- Department of Hematology, Erasmus University Medical Center, Rotterdam
| | | | - Anton W Langerak
- Department of Immunology, Erasmus University Medical Center, Rotterdam.
| |
Collapse
|
16
|
Hasanpour Segherlou Z, Nouri-Vaskeh M, Noroozi Guilandehi S, Baghbanzadeh A, Zand R, Baradaran B, Zarei M. GDF-15: Diagnostic, prognostic, and therapeutic significance in glioblastoma multiforme. J Cell Physiol 2021; 236:5564-5581. [PMID: 33580506 DOI: 10.1002/jcp.30289] [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: 10/10/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the commonest primary malignant brain tumor and has a remarkably weak prognosis. According to the aggressive form of GBM, understanding the accurate molecular mechanism associated with GBM pathogenesis is essential. Growth differentiation factor 15 (GDF-15) belongs to transforming growth factor-β superfamily with important roles to control biological processes. It affects cancer growth and progression, drug resistance, and metastasis. It also can promote stemness in many cancers, and also can stress reactions control, bone generation, hematopoietic growth, adipose tissue performance, and body growth, and contributes to cardiovascular disorders. The role GDF-15 to develop and progress cancer is complicated and remains unclear. GDF-15 possesses tumor suppressor properties, as well as an oncogenic effect. GDF-15 antitumorigenic and protumorigenic impacts on tumor development are linked to the cancer type and stage. However, the GDF-15 signaling and mechanism have not yet been completely identified because of no recognized cognate receptor.
Collapse
Affiliation(s)
| | - Masoud Nouri-Vaskeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Zand
- Department of Neurology, Geisinger Health System, Danville, Pennsylvania, USA
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Zarei
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Li K, Zhang Z, Mei Y, Yang Q, Qiao S, Ni C, Yao Y, Li X, Li M, Wei D, Fu W, Guo X, Huang X, Yang H. Metallothionein-1G suppresses pancreatic cancer cell stemness by limiting activin A secretion via NF-κB inhibition. Theranostics 2021; 11:3196-3212. [PMID: 33537082 PMCID: PMC7847690 DOI: 10.7150/thno.51976] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Resistance to chemotherapy is a long-standing problem in the management of cancer, and cancer stem cells are regarded as the main source of this resistance. This study aimed to investigate metallothionein (MT)-1G involvement in the regulation of cancer stemness and provide a strategy to overcome chemoresistance in pancreatic ductal adenocarcinoma (PDAC). Methods: MT1G was identified as a critical factor related with gemcitabine resistance in PDAC cells by mRNA microarray. Its effects on PDAC stemness were evaluated through sphere formation and tumorigenicity. LC-MS/MS analysis of conditional medium revealed that activin A, a NF-κB target, was a major protein secreted from gemcitabine resistant PDAC cells. Both loss-of-function and gain-of-function approaches were used to validate that MT1G inhibited NF-κB-activin A pathway. Orthotopic pancreatic tumor model was employed to explore the effects on gemcitabine resistance with recombinant follistatin to block activin A. Results: Downregulation of MT1G due to hypermethylation of its promoter is related with pancreatic cancer stemness. Secretome analysis revealed that activin A, a NF-κB target, was highly secreted by drug resistant cells. It promotes pancreatic cancer stemness in Smad4-dependent or independent manners. Mechanistically, MT1G negatively regulates NF-κB signaling and promotes the degradation of NF-κB p65 subunit by enhancing the expression of E3 ligase TRAF7. Blockade of activin A signaling with follistatin could overcome gemcitabine resistance. Conclusions: MT1G suppresses PDAC stemness by limiting activin A secretion via NF-κB inhibition. The blockade of the activin A signaling with follistatin may provide a promising therapeutic strategy for overcoming gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
- Kai Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhicheng Zhang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yu Mei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Qingzhu Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Shupei Qiao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Cheng Ni
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yao Yao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xinyuan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Mengmeng Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Dongdong Wei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Wangjun Fu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xuefei Guo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xuemei Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
18
|
Tanimura Y, Fukui T, Horitani S, Matsumoto Y, Miyamoto S, Suzuki R, Tanaka T, Tomiyama T, Ikeura T, Ando Y, Nishio A, Okazaki K. Long-term model of colitis-associated colorectal cancer suggests tumor spread mechanism and nature of cancer stem cells. Oncol Lett 2020; 21:7. [PMID: 33240413 DOI: 10.3892/ol.2020.12268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Although chemical-induced animal models of colorectal cancer (CRC) suggest a lot about the disease, more efforts are required to establish metastasis models. Azoxymethane (AOM) and dextran sodium sulfate (DSS)-treated (AOM/DSS) Crl:CD-1 mice were sacrificed after 10 or 20 weeks in our previous study, and most colon tumors exhibited intramucosal adenocarcinomas. Our observations were extended until 30 weeks to study a colitis-associated advanced CRC mouse model, and explore whether linker threonine-phosphorylated Smad2/3 (pSmad2/3L-Thr) immunostaining-positive cells were involved in the progressive course of colitis-associated CRC as cancer stem cells. AOM/DSS mice were sacrificed at 10, 20 and 30 weeks after AOM administration. Following the histopathological analysis, immunohistochemical staining was performed for the following markers: CD34, podoplanin, β-catenin, E-cadherin, Ki67, Bmi1 and pSmad2/3L-Thr. Compared with AOM/DSS mice at 10 and 20 weeks, submucosal tumor infiltration and tumor invasion into vessels were markedly increased at 30 weeks. In the parts of colon tumors from AOM/DSS mice, particularly in mice at 30 weeks, the positive signal of E-cadherin was clearly reduced in the cell membranes. The percentage of Ki67-positive tumor cells in mucosal areas of AOM/DSS mice was higher than that in the sites of submucosal infiltration. In mucosal areas of colon tumors, pSmad2/3L-Thr-positive cells were scattered among tumor cells. At sites of submucosal infiltration and vessel invasion of these tumors, pSmad2/3L-Thr-positive cells were also observed among tumor cells. In colon tumors from AOM/DSS mice at 30 weeks, the percentage of pSmad2/3L-Thr-positive cells among the nuclear β-catenin-positive tumor cells was higher than that among the cytoplasmic β-catenin-positive tumor cells. For both non-neoplastic and neoplastic epithelial cells, pSmad2/3L-Thr-positive cells exhibited immunohistochemical co-localization with Bmi1. The present study developed an advanced CRC mouse model that exhibited tumor infiltration into the submucosa and invasion into vessels. The present study re-confirmed the theory that pSmad2/3L-Thr-positive cells may be cancer stem cells.
Collapse
Affiliation(s)
- Yuji Tanimura
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Toshiro Fukui
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Shunsuke Horitani
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Yasushi Matsumoto
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Sachi Miyamoto
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Ryo Suzuki
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Toshihiro Tanaka
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Takashi Tomiyama
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Tsukasa Ikeura
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Yugo Ando
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Akiyoshi Nishio
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Kazuichi Okazaki
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
19
|
Lee CH, Choi Y, Cho H, Bang IH, Hao L, Lee SO, Jeon R, Bae EJ, Park BH. Histone deacetylase 8 inhibition alleviates cholestatic liver injury and fibrosis. Biochem Pharmacol 2020; 183:114312. [PMID: 33130126 DOI: 10.1016/j.bcp.2020.114312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022]
Abstract
Cholestasis is a pathological condition involving blockage of bile flow that results in hepatotoxicity, inflammation, and fibrosis. Although recent studies have shown that histone deacetylases (HDACs) are involved in the progression of fibrosis in various organs, the role of HDAC8 on liver fibrosis has until now remained unexplored. This study presents a newly-synthesized, selective HDAC8 inhibitor SPA3014 composed of a vinyl disulfide-sulfoxide core, and evaluates its therapeutic efficacy against cholestatic liver injury and fibrosis in bile duct-ligated (BDL) mice. We first observed the increase in HDAC8 protein levels in mice with BDL and patients with cholestatic liver disease. Mice with BDL that were pretreated with SPA3014 had lower liver damage and fibrosis, based on gross examination, histopathologic findings, and biochemical analyses, than did vehicle-treated mice. Studies with LX-2 human hepatic stellate cells showed that SPA3014 exerted protective effects by inhibiting TGF-β-mediated activation of MAPK-Smad2/3 and JAK2-STAT3 pathways and by upregulating PPARγ expression. Overall, these results strongly suggest that HDAC8 inhibition constitutes a new therapeutic strategy for treatment of cholestatic liver injury.
Collapse
Affiliation(s)
- Chang Hun Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Yunjung Choi
- Division of Rheumatology, Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Hyewon Cho
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - In Hyuk Bang
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Lihua Hao
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Seung-Ok Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Raok Jeon
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| | - Eun Ju Bae
- College of Pharmacy, Chonbuk National University, Jeonju, Jeonbuk 54896, Republic of Korea.
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea.
| |
Collapse
|
20
|
Decker JT, Kandagatla P, Wan L, Bernstein R, Ma JA, Shea LD, Jeruss JS. Cyclin E overexpression confers resistance to trastuzumab through noncanonical phosphorylation of SMAD3 in HER2+ breast cancer. Cancer Biol Ther 2020; 21:994-1004. [PMID: 33054513 DOI: 10.1080/15384047.2020.1818518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The efficacy of trastuzumab, a treatment for HER2+ breast cancer, can be limited by the development of resistance. Cyclin E (CCNE) overexpression has been implicated in trastuzumab resistance. We sought to uncover a potential mechanism for this trastuzumab resistance and focused on a model of CCNE overexpressing HER2+ breast cancer and noncanonical phosphorylation of the TGF-β signaling protein, SMAD3. Network analysis of transcriptional activity in a HER2+, CCNE overexpressing, trastuzumab-resistant cell line (BT474R2) identified decreased SMAD3 activity was associated with treatment resistance. Immunoblotting showed SMAD3 expression was significantly downregulated in BT474R2 cells (p < .01), and noncanonical phosphorylation of SMAD3 was increased in these CCNE-overexpressing cells. Also, in response to CDK2 inhibition, expression patterns linked to restored canonical SMAD3 signaling, including decreased cMyc and increased cyclin-dependent inhibitor, p15, were identified. The BT474R2 cell line was modified through overexpression of SMAD3 (BT474R2-SMAD3), a mutant construct resistant to CCNE-mediated noncanonical phosphorylation of SMAD3 (BT474R2-5M), and a control (BT474R2-Blank). In vitro studies examining the response to trastuzumab showed increased sensitivity to treatment for BT474R2-5M cells. These findings were then validated in NSG mice inoculated with BT474R2-5M cells or BT474R2 control cells. After treatment with trastuzumab, the NSG mice inoculated with BT474R2-5M cells developed significantly lower tumor volumes (p < .001), when compared to mice inoculated with BT474R2 cells. Taken together, these results indicate that for patients with HER2+ breast cancer, a mechanism of CCNE-mediated trastuzumab resistance, regulated through noncanonical SMAD3 phosphorylation, could be treated with CDK2 inhibition to help enhance the efficacy of trastuzumab therapy.
Collapse
Affiliation(s)
- Joseph T Decker
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Pridvi Kandagatla
- Department of Surgery, Henry Ford Health System , Detroit, MI, USA.,Department of Surgery, University of Michigan , Ann Arbor, MI, USA
| | - Lei Wan
- Department of Surgery, University of Michigan , Ann Arbor, MI, USA
| | - Regan Bernstein
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Jeffrey A Ma
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA.,Department of Surgery, University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
21
|
Cao X, Dang L, Zheng X, Lu Y, Lu Y, Ji R, Zhang T, Ruan X, Zhi J, Hou X, Yi X, Li MJ, Gu T, Gao M, Zhang L, Chen Y. Targeting Super-Enhancer-Driven Oncogenic Transcription by CDK7 Inhibition in Anaplastic Thyroid Carcinoma. Thyroid 2019; 29:809-823. [PMID: 30924726 DOI: 10.1089/thy.2018.0550] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Anaplastic thyroid carcinoma (ATC) is one of the most aggressive malignancies, with no effective treatment currently available. The molecular mechanisms of ATC carcinogenesis remain poorly understood. The objective of this study was to investigate the mechanisms and functions of super-enhancer (SE)-driven oncogenic transcriptional addiction in the progression of ATC and identify new drug targets for ATC treatments. Methods: High-throughput chemical screening was performed to identify new drugs inhibiting ATC cell growth. Cell viability assay, colony formation analysis, cell-cycle analysis, and animal study were used to examine the effects of drug treatments on ATC progression. Chromatin immunoprecipitation sequencing was conducted to establish a SE landscape of ATC. Integrative analysis of RNA sequencing, chromatin immunoprecipitation sequencing, and CRISPR/Cas9-mediated gene editing was used to identify THZ1 target genes. Drug combination analysis was performed to assess drug synergy. Patient samples were analyzed to evaluate candidate biomarkers of prognosis in ATC. Results: THZ1, a covalent inhibitor of cyclin-dependent kinase 7 (CDK7), was identified as a potent anti-ATC compound by high-throughput chemical screening. ATC cells, but not papillary thyroid carcinoma cells, are exceptionally sensitive to CDK7 inhibition. An integrative analysis of both gene expression profiles and SE features revealed that the SE-mediated oncogenic transcriptional amplification mediates the vulnerability of ATC cells to THZ1 treatment. Combining this integrative analysis with functional assays led to the discovery of a number of novel cancer genes of ATC, including PPP1R15A, SMG9, and KLF2. Inhibition of PPP1R15A with Guanabenz or Sephin1 greatly suppresses ATC growth. Significantly, the expression level of PPP1R15A is correlated with CDK7 expression in ATC tissue samples. Elevated expression of PPP1R15A and CDK7 are both associated with poor clinical prognosis in ATC patients. Importantly, CDK7 or PPP1R15A inhibition sensitizes ATC cells to conventional chemotherapy. Conclusions: Taken together, these findings demonstrate transcriptional addiction in ATC pathobiology and identify CDK7 and PPP1R15A as potential biomarkers and therapeutic targets for ATC.
Collapse
Affiliation(s)
- Xinyi Cao
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Lin Dang
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Xiangqian Zheng
- 2 Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, P.R. China
| | - Yi Lu
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Yumei Lu
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Rongjie Ji
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Tianye Zhang
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Xianhui Ruan
- 2 Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, P.R. China
| | - Jingtai Zhi
- 2 Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, P.R. China
| | - Xiukun Hou
- 2 Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, P.R. China
| | - Xianfu Yi
- 3 School of Biomedical Engineering, Tianjin Medical University, Tianjin, P.R. China
| | - Mulin Jun Li
- 4 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P.R. China
| | - Tingyu Gu
- 5 Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Ming Gao
- 2 Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, P.R. China
| | - Lirong Zhang
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
| | - Yupeng Chen
- 1 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, P.R. China
- 2 Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, P.R. China
| |
Collapse
|
22
|
Fuchs C, Medici G, Trazzi S, Gennaccaro L, Galvani G, Berteotti C, Ren E, Loi M, Ciani E. CDKL5 deficiency predisposes neurons to cell death through the deregulation of SMAD3 signaling. Brain Pathol 2019; 29:658-674. [PMID: 30793413 DOI: 10.1111/bpa.12716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
CDKL5 deficiency disorder (CDD) is a rare encephalopathy characterized by early onset epilepsy and severe intellectual disability. CDD is caused by mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene, a member of a highly conserved family of serine-threonine kinases. Only a few physiological substrates of CDKL5 are currently known, which hampers the discovery of therapeutic strategies for CDD. Here, we show that SMAD3, a primary mediator of TGF-β action, is a direct phosphorylation target of CDKL5 and that CDKL5-dependent phosphorylation promotes SMAD3 protein stability. Importantly, we found that restoration of the SMAD3 signaling through TGF-β1 treatment normalized defective neuronal survival and maturation in Cdkl5 knockout (KO) neurons. Moreover, we demonstrate that Cdkl5 KO neurons are more vulnerable to neurotoxic/excitotoxic stimuli. In vivo treatment with TGF-β1 prevents increased NMDA-induced cell death in hippocampal neurons from Cdkl5 KO mice, suggesting an involvement of the SMAD3 signaling deregulation in the neuronal susceptibility to excitotoxic injury of Cdkl5 KO mice. Our finding reveals a new function for CDKL5 in maintaining neuronal survival that could have important implications for susceptibility to neurodegeneration in patients with CDD.
Collapse
Affiliation(s)
- Claudia Fuchs
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giorgio Medici
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Laura Gennaccaro
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Galvani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisa Ren
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Manuela Loi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Lappas M. Identification of SMAD3 as a Novel Mediator of Inflammation in Human Myometrium In Vitro. Mediators Inflamm 2018; 2018:3140420. [PMID: 30363688 PMCID: PMC6180979 DOI: 10.1155/2018/3140420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 02/01/2023] Open
Abstract
Preterm birth remains the primary cause of early neonatal death and is a major determinant for long-term health consequences. Aberrant intrauterine inflammation and infection are known to augment the synthesis of proinflammatory cytokines and induce uterine contractions, which can subsequently lead to preterm birth. The transforming growth factor-β (TGF-β) superfamily members regulate numerous cellular processes through the activation of intracellular mediators known as mothers against decapentaplegic homolog (SMADs). Studies in nongestational tissues have shown that SMAD3 plays a role in immune regulation and inflammation; however, its role in human labour remains unknown. Thus, the present study aimed at (i) characterising the expression of SMAD3 in the human myometrium; (ii) determining the effect of bacterial and viral products and proinflammatory cytokines on SMAD3 transcriptional activity in primary human myometrial cells; and (iii) investigating the effect of SMAD3 siRNA knockdown on the production of prolabour mediators in primary human myometrial cells. Phosphorylated (i.e., active) SMAD3 protein expression was lower in the myometrium after spontaneous term labour compared to the myometrium from nonlabouring women. Using a luciferase assay, the proinflammatory cytokines IL-1β and TNF, and viral analogue polyinosinic : polycytidylic acid (poly(I : C)) significantly reduced SMAD3 transcriptional activity in human primary myometrial cells. Loss-of-function studies found that SMAD3 knockdown in myometrial cells significantly increased IL-1β- and poly(I : C)-induced proinflammatory cytokines (IL-1A, IL-6), chemokines (IL-8, MCP-1), the adhesion molecule ICAM-1, COX-2 mRNA expression, and subsequent PGF2α release. In conclusion, SMAD3 deficiency is associated with increased production of proinflammatory and prolabour mediators in the human myometrium.
Collapse
Affiliation(s)
- Martha Lappas
- Obstetrics, Nutrition, and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
24
|
Huang F, Shi Q, Li Y, Xu L, Xu C, Chen F, Wang H, Liao H, Chang Z, Liu F, Zhang XHF, Feng XH, Han JDJ, Luo S, Chen YG. HER2/EGFR-AKT Signaling Switches TGFβ from Inhibiting Cell Proliferation to Promoting Cell Migration in Breast Cancer. Cancer Res 2018; 78:6073-6085. [PMID: 30171053 DOI: 10.1158/0008-5472.can-18-0136] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/15/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022]
Abstract
TGFβ signaling inhibits cell proliferation to block cancer initiation, yet it also enhances metastasis to promote malignancy during breast cancer development. The mechanisms underlying these differential effects are still unclear. Here, we report that HER2/EGFR signaling switches TGFβ function in breast cancer cells from antiproliferation to cancer promotion. Inhibition of HER2/EGFR activity attenuated TGFβ-induced epithelial-mesenchymal transition and migration but enhanced the antiproliferative activity of TGFβ. Activation of HER2/EGFR induced phosphorylation of Smad3 at Ser208 of the linker region through AKT, which promoted the nuclear accumulation of Smad3 and subsequent expression of the genes related to EMT and cell migration. In contrast, HER2/EGFR signaling had no effects on the nuclear localization of Smad2. Knockdown of Smad3, but not Smad2, blocked TGFβ-induced breast cancer cell migration. We observed a positive correlation between the nuclear localization of Smad3 and HER2 activation in advanced human breast cancers. Our results demonstrate a key role for HER2/EGFR in differential regulation of Smad3 activity to shift TGFβ function from antitumorigenic to protumorigenic during breast cancer development.Significance: TGFβ signaling can shift from inhibiting to promoting breast cancer development via HER2/EGFR AKT-mediated phosphorylation of Smad3 at S208, enhancing its nuclear accumulation and upregulation of EMT-related genes.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/21/6073/F1.large.jpg Cancer Res; 78(21); 6073-85. ©2018 AACR.
Collapse
Affiliation(s)
- Fei Huang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiaoni Shi
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuzhen Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Linlin Xu
- The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Chi Xu
- Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fenfang Chen
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Hongwei Liao
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zai Chang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Fang Liu
- Center for Advanced Biotechnology and Medicine, Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing-Dong J Han
- Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shiwen Luo
- The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
25
|
Tapella L, Cerruti M, Biocotino I, Stevano A, Rocchio F, Canonico PL, Grilli M, Genazzani AA, Lim D. TGF-β2 and TGF-β3 from cultured β-amyloid-treated or 3xTg-AD-derived astrocytes may mediate astrocyte-neuron communication. Eur J Neurosci 2018; 47:211-221. [DOI: 10.1111/ejn.13819] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Matteo Cerruti
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Isabella Biocotino
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Alessio Stevano
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Francesca Rocchio
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| |
Collapse
|
26
|
Li J, Zou K, Yu L, Zhao W, Lu Y, Mao J, Wang B, Wang L, Fan S, Song B, Li L. MicroRNA-140 Inhibits the Epithelial-Mesenchymal Transition and Metastasis in Colorectal Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 10:426-437. [PMID: 29499953 PMCID: PMC5862396 DOI: 10.1016/j.omtn.2017.12.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/30/2017] [Accepted: 12/30/2017] [Indexed: 12/21/2022]
Abstract
MicroRNA-140, a cartilage-specific microRNA, has recently been implicated in the cancer progression. However, the comprehensive role of miR-140 in the invasion and metastasis of colorectal cancer (CRC) is still not fully understood. In this study, we confirmed that miR-140 downregulates SMAD family member 3 (Smad3), which is a key downstream effector of the TGF-β signaling pathway, at the translational level in the CRC cell lines. Ectopic expression of miR-140 inhibits the process of epithelial-mesenchymal transition (EMT), at least partially through targeting Smad3, and induces the suppression of migratory and invasive capacities of CRC cells in vitro. miR-140 also attenuates CRC cell proliferation possibly via downregulating Samd3. Furthermore, overexpression of miR-140 inhibits the tumor formation and metastasis of CRC in vivo, and silenced Smad3 has the similar effect. Additionally, miR-140 expression is decreased in the clinical primary CRC specimens and appears as a progressive reduction in the metastatic specimens, whereas Smad3 is overexpressed in the CRC samples. Taken together, our findings suggest that miR-140 might be a key suppressor of CRC progression and metastasis through inhibiting EMT process by targeting Smad3. miR-140 may represent a novel candidate for CRC treatment.
Collapse
Affiliation(s)
- Jiazhi Li
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Kun Zou
- Department of Oncology Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Lihui Yu
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Wenyue Zhao
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Ying Lu
- Department of Pathology, Dalian Medical University, Dalian 116044, China; Teaching Laboratory of Morphology, Dalian Medical University, Dalian 116044, China
| | - Jun Mao
- Department of Pathology, Dalian Medical University, Dalian 116044, China; The Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Bo Wang
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Lu Wang
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Shujun Fan
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Bo Song
- Department of Pathology, Dalian Medical University, Dalian 116044, China; Teaching Laboratory of Morphology, Dalian Medical University, Dalian 116044, China.
| | - Lianhong Li
- Department of Pathology, Dalian Medical University, Dalian 116044, China; The Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
27
|
Synergistic effect of eribulin and CDK inhibition for the treatment of triple negative breast cancer. Oncotarget 2017; 8:83925-83939. [PMID: 29137393 PMCID: PMC5663565 DOI: 10.18632/oncotarget.20202] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/12/2017] [Indexed: 01/22/2023] Open
Abstract
Activation of CDK2 in triple negative breast cancer (TNBC) can contribute to non-canonical phosphorylation of a TGFβ signaling component, Smad3, promoting cell proliferation and migration. Inhibition of CDK2 was shown to decrease breast cancer oncogenesis. Eribulin chemotherapy was used effectively in the treatment of TNBC. To this end, we tested therapeutic efficacy of a novel CDK2/9 inhibitor, CYC065, eribulin, and the combination of CYC065 and eribulin in 3 different TNBC cell lines, and an in vivo xenograft model. Specifically, we characterized cell proliferation, apoptosis, migration, cell cycle associated protein expression, treatment-related transcription factor activity, and tumor growth in TNBC. Treatment with CYC065 and eribulin in combination had a superior effect on decreasing cell proliferation, inducing apoptosis, and inhibiting migration in TNBC cell lines in vitro. Combination therapy inhibited non-canonical Smad3 phosphorylation at the T179 site in the protein linker region, and resulted in increased p15 and decreased c-myc expression. In a transcription factor array, combination treatment significantly increased activity of AP1 and decreased activity of factors including NFκB, SP1, E2F, and SMAD3. In an in vivo xenograft model of TNBC, individual and combination treatments resulted in a decrease in both tumor volume and mitotic indices. Taken together, these studies highlight the potential of this novel drug combination, CYC065 and eribulin, to suppress the growth of TNBC cells in vitro and in vivo, warranting further clinical investigation.
Collapse
|
28
|
Smad2/3 Linker Phosphorylation Is a Possible Marker of Pancreatic Stem/Progenitor Cells in the Regenerative Phase of Acute Pancreatitis. Pancreas 2017; 46:605-613. [PMID: 28099259 DOI: 10.1097/mpa.0000000000000759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The aims of this study are to characterize cell proliferation and differentiation during regeneration after pancreatitis and pancreatic buds during development to evaluate the role of Smad2/3, phosphorylated at the specific linker threonine residues (pSmad2/3L-Thr) in positive cells. METHODS Male C57BL/6 mice received hourly intraperitoneal injections of cerulein and were analyzed after induced pancreatitis. Pancreatitis-affected tissue sections and pancreatic buds were immunostained for pSmad2/3L-Thr, with other markers thought to be stem/progenitor markers of the pancreas. RESULTS pSmad2/3L-Thr immunostaining-positive cells increased as the pancreatitis progressed. The expression of pSmad2/3L-Thr was seen in acinar cells and ductlike tubular complexes. These results suggest that pSmad2/3L-Thr is expressed during acinar-ductal metaplasia. Immunohistochemical colocalization of pSmad2/3L-Thr with Ki67 was never observed. pSmad2/3L-Thr-positive cells may remain in an undifferentiated state. During the pancreatic development process, pSmad2/3L-Thr was expressed as other markers. pSmad2/3L-Thr develops in duct structure of the undifferentiated cell population in the last part of viviparity that acinar structure is formed clearly. CONCLUSIONS pSmad2/3L-Thr expression occurs during acinar-ductal metaplasia after pancreatitis and may represent the contribution of stem cells and/or progenitor cells to the differentiation of the pancreas.
Collapse
|
29
|
Lnc-ATB contributes to gastric cancer growth through a MiR-141-3p/TGFβ2 feedback loop. Biochem Biophys Res Commun 2017; 484:514-521. [PMID: 28115163 DOI: 10.1016/j.bbrc.2017.01.094] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 12/17/2022]
Abstract
The long noncoding RNA (lncRNA) ATB is an important regulator in human tumors. Here, we aimed to investigate the potential molecular mechanisms of lnc-ATB in gastric cancer (GC) tumorigenesis. RT-qPCR analysis was used to detect lnc-ATB expression level in 20 pairs of gastric cancer tissues and adjacent normal gastric mucosa tissues (ANTs). Moreover, the biological role of lnc-ATB was determined in vitro. We found that lnc-ATB was significantly upregulated in GC tissues compared to lnc-ATB expression in ANTs. These high lnc-ATB expression levels predicted poor prognosis in GC patients. Low levels of lnc-ATB inhibited GC cell proliferation and cell cycle arrest in vitro. Lnc-ATB was found to directly bind miR-141-3p. Moreover, TGF-β actives lnc-ATB and TGF-β2 directly binds mir-141-3p. Finally, we demonstrated that lnc-ATB fulfilled its oncogenic roles in a ceRNA-mediated manner. Our study suggests that lnc-ATB promotes tumor progression by interacting with miR-141-3p and that Lnc-ATB may be a valuable prognostic predictor for GC. In conclusion, the positive feedback loop of lnc-ATB/miR-141-3p/TGF-β2 may be a potential therapeutic target for the treatment of GC.
Collapse
|
30
|
Zhang L, Cheng X, Gao Y, Zhang C, Bao J, Guan H, Yu H, Lu R, Xu Q, Sun Y. Curcumin inhibits metastasis in human papillary thyroid carcinoma BCPAP cells via down-regulation of the TGF-β/Smad2/3 signaling pathway. Exp Cell Res 2016; 341:157-65. [PMID: 26826337 DOI: 10.1016/j.yexcr.2016.01.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/12/2016] [Accepted: 01/16/2016] [Indexed: 01/07/2023]
Abstract
Thyroid cancers usually possess a good prognosis while the risks of recurrence and metastasis turn out to be a disturbing issue. Curcumin [bis(4-hydroxy-3-methoxy-phenyl)-1,6-heptadiene-3,5-dione] is a natural polyphenolic compound mainly found in turmeric (Curcuma longa). Our previous studies have demonstrated that curcumin showed proliferation-inhibitory and apoptosis-inducing effects on K1 papillary thyroid cancer cells. However, the mechanism underlying the inhibition effects of curcumin on thyroid cancer cells remains unclear. Herein, we demonstrated that curcumin remarkably increased the expression of the epithelial marker E-cadherin and repressed the expression of the mesenchymal marker vimentin in human papillary thyroid carcinoma BCPAP cells. Curcumin also suppressed multiple metastatic steps of BCPAP cells, including cell attachment, spreading as well as migration. In addition, the transcription, secretion and activation of matrix metalloproteinases (MMPs) induced by transforming growth factor-β1 (TGF-β1) in BCPAP cells were mitigated upon curcumin treatment. Further evidence showed that curcumin decreased TGF-β1-mediated phosphorylation of Smad2 and Smad3. These results revealed that curcumin inhibited the TGF-β1-induced epithelial-mesenchymal transition (EMT) via down-regulation of Smad2/3 signaling pathways. Our findings provide new evidence that the anti-metastatic and anti-EMT activities of curcumin may contribute to the development of chemo-preventive agents for thyroid cancer treatment.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China; Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Xian Cheng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yanyan Gao
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Chiyu Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiandong Bao
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Haixia Guan
- Department of Endocrinology & Metabolism and Institute of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huixin Yu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Rongrong Lu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
31
|
Lin RL, Zhao LJ. Mechanistic basis and clinical relevance of the role of transforming growth factor-β in cancer. Cancer Biol Med 2016; 12:385-93. [PMID: 26779375 PMCID: PMC4706525 DOI: 10.7497/j.issn.2095-3941.2015.0015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a key factor in cancer development and progression. TGF-β can suppress tumorigenesis by inhibiting cell cycle progression and stimulating apoptosis in early stages of cancer progression. However, TGF-β can modulate cancer-related processes, such as cell invasion, distant metastasis, and microenvironment modification that may be used by cancer cells to their advantage in late stages. Corresponding mechanisms include angiogenesis promotion, anti-tumor immunity suppression, and epithelial-to-mesenchymal transition (EMT) induction. The correlation between TGF-β expression and cancer prognosis has also been extensively investigated. Results suggest that TGF-β pathway can be targeted to treat cancer; as such, the feasibility of this treatment is investigated in clinical trials.
Collapse
Affiliation(s)
- Run-Long Lin
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Lu-Jun Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
32
|
Yu J, Chen C, Xu T, Yan M, Xue B, Wang Y, Liu C, Zhong T, Wang Z, Meng X, Hu D, Yu X. Pseudolaric acid B activates autophagy in MCF-7 human breast cancer cells to prevent cell death. Oncol Lett 2016; 11:1731-1737. [PMID: 26998069 DOI: 10.3892/ol.2016.4103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/24/2015] [Indexed: 01/08/2023] Open
Abstract
Pseudolaric acid B (PAB) has been demonstrated to exert antitumor effects in MCF-7 human breast cancer cells. The present study aimed to investigate the mechanism of resistance to PAB-induced cell death. Following incubation with 4 µM of PAB for 3 days, the majority of MCF-7 cells became senescent, while some retained the same morphology as control cells, as assessed using a senescence detection kit. Additionally, 36 h of treatment with 4 µM of PAB increased the positive staining of autophagy markers, as shown by monodansylcadaverine and acridine orange staining. Western blot analysis indicated that this treatment also increased expression of the autophagy-related proteins Beclin-1 and microtubule-associated protein 1 light chain 3. Furthermore, treatment with PAB and the autophagy inhibitor 3-methyl adenine significantly decreased the ratio of autophagy, as assessed by flow cytometric analysis of monodansylcadaverine staining density (P<0.001), and increased the ratio of cell death, as assessed by MTT analysis (P<0.001). This indicated that autophagy promotes cell survival as a resistance mechanism to PAB treatment. Additionally, the present study demonstrated that PAB treatment did not affect the mitochondrial membrane potential, which may be related to autophagy. Increased Bcl-2 expression may explain why PAB did not affect the mitochondrial membrane potential. A Bcl-2 binding test demonstrated that PAB treatment inhibits the binding of Bcl-2 and Beclin-1, which may free Beclin-1 to participate in autophagy. Therefore, the present study demonstrated that autophagy may be activated by PAB treatment in human breast cancer MCF-7 cells, contributing to resistance to cell death.
Collapse
Affiliation(s)
- Jinghua Yu
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chunhai Chen
- Department of Acupunture, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Tianyang Xu
- Department of Drug Analysis and Analytical Chemistry, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Minghui Yan
- Department of Biomedical Engineering, College of Pharmacy, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Bianbian Xue
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ying Wang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chunyu Liu
- Department of Acupunture, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Ting Zhong
- Department of Medicinal Chemistry, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Zengyan Wang
- Department of Internal Medicine, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xianying Meng
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Donghua Hu
- Department of Medicinal Chemistry, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Xiaofang Yu
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
Aguado BA, Wu JJ, Azarin SM, Nanavati D, Rao SS, Bushnell GG, Medicherla CB, Shea LD. Secretome identification of immune cell factors mediating metastatic cell homing. Sci Rep 2015; 5:17566. [PMID: 26634905 PMCID: PMC4669442 DOI: 10.1038/srep17566] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/02/2015] [Indexed: 12/25/2022] Open
Abstract
Metastatic cell homing is a complex process mediated in part by diffusible factors secreted from immune cells found at a pre-metastatic niche. We report on connecting secretomics and TRanscriptional Activity CEll aRray (TRACER) data to identify functional paracrine interactions between immune cells and metastatic cells as novel mediators of homing. Metastatic breast cancer mouse models were used to generate a diseased splenocyte conditioned media (D-SCM) containing immune cell secreted factors. MDA-MB-231 metastatic cell activity including cell invasion, migration, transendothelial migration, and proliferation were increased in D-SCM relative to control media. Our D-SCM secretome analysis yielded 144 secreted factor candidates that contribute to increased metastatic cell activity. The functional mediators of homing were identified using MetaCore software to determine interactions between the immune cell secretome and the TRACER-identified active transcription factors within metastatic cells. Among the 5 candidate homing factors identified, haptoglobin was selected and validated in vitro and in vivo as a key mediator of homing. Our studies demonstrate a novel systems biology approach to identify functional signaling factors associated with a cellular phenotype, which provides an enabling tool that complements large-scale protein identification provided by proteomics.
Collapse
Affiliation(s)
- Brian A Aguado
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA.,Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Jia J Wu
- Interdepartmental Biological Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dhaval Nanavati
- Proteomics Core Facility, Northwestern University, Chicago, IL 60611, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | | | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.,Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.,Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
34
|
Abstract
Transforming growth factor-β (TGF-β) is a key factor in cancer development and progression. TGF-β can suppress tumorigenesis by inhibiting cell cycle progression and stimulating apoptosis in early stages of cancer progression. However, TGF-β can modulate cancer-related processes, such as cell invasion, distant metastasis, and microenvironment modification that may be used by cancer cells to their advantage in late stages. Corresponding mechanisms include angiogenesis promotion, anti-tumor immunity suppression, and epithelial-to-mesenchymal transition (EMT) induction. The correlation between TGF-β expression and cancer prognosis has also been extensively investigated. Results suggest that TGF-β pathway can be targeted to treat cancer; as such, the feasibility of this treatment is investigated in clinical trials.
Collapse
Affiliation(s)
- Run-Long Lin
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Lu-Jun Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
35
|
Fabian CJ, Kimler BF, Phillips TA, Box JA, Kreutzjans AL, Carlson SE, Hidaka BH, Metheny T, Zalles CM, Mills GB, Powers KR, Sullivan DK, Petroff BK, Hensing WL, Fridley BL, Hursting SD. Modulation of Breast Cancer Risk Biomarkers by High-Dose Omega-3 Fatty Acids: Phase II Pilot Study in Premenopausal Women. Cancer Prev Res (Phila) 2015; 8:912-21. [PMID: 26438592 PMCID: PMC6053670 DOI: 10.1158/1940-6207.capr-14-0335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Higher intakes of the omega-3 eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) relative to the omega-6 arachidonic acid (AA) have been variably associated with reduced risk of premenopausal breast cancer. The purpose of this pilot trial was to assess feasibility and explore the effects of high-dose EPA and DHA on blood and benign breast tissue risk biomarkers before design of a placebo-controlled phase IIB trial. Premenopausal women with evidence of hyperplasia ± atypia by baseline random periareolar fine needle aspiration were given 1860 mg of EPA + 1500 mg of DHA ethyl esters daily for 6 months. Blood and benign breast tissue were sampled during the same menstrual cycle phase prestudy and a median of 3 weeks after last dose. Additional blood was obtained within 24 hours of last dose. Feasibility, which was predefined as 50% uptake, 85% retention, and 70% compliance, was demonstrated with 46% uptake, 94% completion, and 85% compliance. Cytologic atypia decreased from 77% to 38% (P = 0.002), and Ki-67 from a median of 2.1% to 1.0% (P = 0.021) with an increase in the ratio of EPA + DHA to AA in erythrocyte phospholipids but no change in blood hormones, adipokines, or cytokines. Exploratory breast proteomics assessment showed decreases in several proteins involved in hormone and cytokine signaling with mixed effects on those in the AKT/mTOR pathways. Further investigation of EPA plus DHA for breast cancer prevention in a placebo-controlled trial in premenopausal women is warranted.
Collapse
Affiliation(s)
- Carol J Fabian
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruce F Kimler
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas
| | - Teresa A Phillips
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Jessica A Box
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Amy L Kreutzjans
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas
| | - Brandon H Hidaka
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas
| | - Trina Metheny
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kandy R Powers
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas
| | - Brian K Petroff
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Whitney L Hensing
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Brooke L Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
36
|
Fabian CJ, Kimler BF, Phillips TA, Nydegger JL, Kreutzjans AL, Carlson SE, Hidaka BH, Metheny T, Zalles CM, Mills GB, Powers KR, Sullivan DK, Petroff BK, Hensing WL, Fridley BL, Hursting SD. Modulation of Breast Cancer Risk Biomarkers by High-Dose Omega-3 Fatty Acids: Phase II Pilot Study in Postmenopausal Women. Cancer Prev Res (Phila) 2015; 8:922-31. [PMID: 26276744 PMCID: PMC4596784 DOI: 10.1158/1940-6207.capr-14-0336] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 08/03/2015] [Indexed: 01/05/2023]
Abstract
Associational studies suggest higher intakes/blood levels of the omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) relative to the omega-6 arachidonic acid (AA) are associated with reduced breast cancer risk. We performed a pilot study of high-dose EPA + DHA in postmenopausal women to assess feasibility before initiating a phase IIB prevention trial. Postmenopausal women with cytologic evidence of hyperplasia in their baseline random periareolar fine needle aspiration (RPFNA) took 1,860 mg EPA +1500 mg DHA ethyl esters daily for 6 months. Blood and breast tissue were sampled at baseline and study conclusion for exploratory biomarker assessment, with P values uncorrected for multiple comparisons. Feasibility was predefined as 50% uptake, 80% completion, and 70% compliance. Trial uptake by 35 study entrants from 54 eligible women was 65%, with 97% completion and 97% compliance. Favorable modulation was suggested for serum adiponectin (P = 0.0027), TNFα (P = 0.016), HOMA 2B measure of pancreatic β cell function (P = 0.0048), and bioavailable estradiol (P = 0.039). Benign breast tissue Ki-67 (P = 0.036), macrophage chemoattractant protein-1 (P = 0.033), cytomorphology index score (P = 0.014), and percent mammographic density (P = 0.036) were decreased with favorable effects in a proteomics array for several proteins associated with mitogen signaling and cell-cycle arrest; but no obvious overall effect on proteins downstream of mTOR. Although favorable risk biomarker modulation will need to be confirmed in a placebo-controlled trial, we have demonstrated feasibility for development of high-dose EPA and DHA ethyl esters for primary prevention of breast cancer.
Collapse
Affiliation(s)
- Carol J Fabian
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruce F Kimler
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas
| | - Teresa A Phillips
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Jennifer L Nydegger
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Amy L Kreutzjans
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas
| | - Brandon H Hidaka
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas
| | - Trina Metheny
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kandy R Powers
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas
| | - Brian K Petroff
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Whitney L Hensing
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Brooke L Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
37
|
Suzuki R, Fukui T, Kishimoto M, Miyamoto S, Takahashi Y, Takeo M, Mitsuyama T, Sakaguchi Y, Uchida K, Nishio A, Okazaki K. Smad2/3 linker phosphorylation is a possible marker of cancer stem cells and correlates with carcinogenesis in a mouse model of colitis-associated colorectal cancer. J Crohns Colitis 2015; 9:565-74. [PMID: 25908723 DOI: 10.1093/ecco-jcc/jjv073] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/14/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Epithelial cells affected by somatic mutations undergo transition from a tumour-suppressive to a carcinogenic Smad pathway during sporadic colorectal carcinogenesis, and the specific linker threonine phosphorylation of Smad2/3 in colon epithelial cells indicates stem-like cells. This study extends previous observations to a model of colitis-associated colorectal cancer. METHODS After Crl:CD-1 mice received an administration of azoxymethane [AOM], the mice were given dextran sodium sulfate [DSS] for 7 days. AOM/DSS-treated mice [AOM/DSS mice] were killed at 10 or 20 weeks. After macroscopic observations, a histopathological analysis was conducted. Immunohistochemical staining was performed using the avidin-biotin immunoperoxidase method [pSmad3C-Ser, pSmad3L-Ser, c-Myc] and immunofluorescent methods [Ki67, β-catenin, CDK4, cyclin D1, Sox9, pSmad2/3L-Thr]. RESULTS The colons from AOM/DSS mice were shorter than those from control mice. The number of colon tumours at Week 20 was higher than at Week 10. The inflammation scores for AOM/DSS mice were greater than those for control mice. Immunostaining-positive cells (staining by Ki67, β-catenin [nuclear and cytoplasmic], cyclin D1, and Sox9) were diffusely distributed in colon tumours. The percentage of pSmad3L-Ser-positive cells in colon tumours was higher than in sites of pre-neoplastic colitis, and that in sites of pre-neoplastic colitis was higher than in control mice. pSmad2/3L-Thr-positive cells were sparsely detected around crypt bases in non-neoplastic colon epithelia and at the tops of tumours, and immunohistochemical co-localisation of pSmad2/3L-Thr with Ki67 was not observed. Immunohistochemical co-localisation of pSmad2/3L-Thr with β-catenin and CDK4 was observed. CONCLUSIONS pSmad3L-Ser signalling is an early event in colitis-associated colorectal cancer, and pSmad2/3L-Thr immunostaining-positive cells might be cancer stem cells.
Collapse
Affiliation(s)
- Ryo Suzuki
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Toshiro Fukui
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Masanobu Kishimoto
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Sachi Miyamoto
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Yu Takahashi
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Masahiro Takeo
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Toshiyuki Mitsuyama
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Yutaku Sakaguchi
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Kazushige Uchida
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Akiyoshi Nishio
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Kazuichi Okazaki
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
38
|
Kaklamani VG, Jeruss JS, Hughes E, Siziopikou K, Timms KM, Gutin A, Abkevich V, Sangale Z, Solimeno C, Brown KL, Jones J, Hartman AR, Meservey C, Jovanovic B, Helenowski I, Khan SA, Bethke K, Hansen N, Uthe R, Giordano S, Rosen S, Hoskins K, Von Roenn J, Jain S, Parini V, Gradishar W. Phase II neoadjuvant clinical trial of carboplatin and eribulin in women with triple negative early-stage breast cancer (NCT01372579). Breast Cancer Res Treat 2015; 151:629-38. [DOI: 10.1007/s10549-015-3435-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/20/2022]
|
39
|
Deregulation of the EGFR/PI3K/PTEN/Akt/mTORC1 pathway in breast cancer: possibilities for therapeutic intervention. Oncotarget 2015; 5:4603-50. [PMID: 25051360 PMCID: PMC4148087 DOI: 10.18632/oncotarget.2209] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The EGFR/PI3K/PTEN/Akt/mTORC1/GSK-3 pathway plays prominent roles in malignant transformation, prevention of apoptosis, drug resistance and metastasis. The expression of this pathway is frequently altered in breast cancer due to mutations at or aberrant expression of: HER2, ERalpha, BRCA1, BRCA2, EGFR1, PIK3CA, PTEN, TP53, RB as well as other oncogenes and tumor suppressor genes. In some breast cancer cases, mutations at certain components of this pathway (e.g., PIK3CA) are associated with a better prognosis than breast cancers lacking these mutations. The expression of this pathway and upstream HER2 has been associated with breast cancer initiating cells (CICs) and in some cases resistance to treatment. The anti-diabetes drug metformin can suppress the growth of breast CICs and herceptin-resistant HER2+ cells. This review will discuss the importance of the EGFR/PI3K/PTEN/Akt/mTORC1/GSK-3 pathway primarily in breast cancer but will also include relevant examples from other cancer types. The targeting of this pathway will be discussed as well as clinical trials with novel small molecule inhibitors. The targeting of the hormone receptor, HER2 and EGFR1 in breast cancer will be reviewed in association with suppression of the EGFR/PI3K/PTEN/Akt/mTORC1/GSK-3 pathway.
Collapse
|
40
|
Li Y, Song D, Song Y, Zhao L, Wolkow N, Tobias JW, Song W, Dunaief JL. Iron-induced Local Complement Component 3 (C3) Up-regulation via Non-canonical Transforming Growth Factor (TGF)-β Signaling in the Retinal Pigment Epithelium. J Biol Chem 2015; 290:11918-34. [PMID: 25802332 PMCID: PMC4424331 DOI: 10.1074/jbc.m115.645903] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 03/20/2015] [Indexed: 12/26/2022] Open
Abstract
Dysregulation of iron homeostasis may be a pathogenic factor in age-related macular degeneration (AMD). Meanwhile, the formation of complement-containing deposits under the retinal pigment epithelial (RPE) cell layer is a pathognomonic feature of AMD. In this study, we investigated the molecular mechanisms by which complement component 3 (C3), a central protein in the complement cascade, is up-regulated by iron in RPE cells. Modulation of TGF-β signaling, involving ERK1/2, SMAD3, and CCAAT/enhancer-binding protein-δ, is responsible for iron-induced C3 expression. The differential effects of spatially distinct SMAD3 phosphorylation sites at the linker region and at the C terminus determined the up-regulation of C3. Pharmacologic inhibition of either ERK1/2 or SMAD3 phosphorylation decreased iron-induced C3 expression levels. Knockdown of SMAD3 blocked the iron-induced up-regulation and nuclear accumulation of CCAAT/enhancer-binding protein-δ, a transcription factor that has been shown previously to bind the basic leucine zipper 1 domain in the C3 promoter. We show herein that mutation of this domain reduced iron-induced C3 promoter activity. In vivo studies support our in vitro finding of iron-induced C3 up-regulation. Mice with a mosaic pattern of RPE-specific iron overload demonstrated co-localization of iron-induced ferritin and C3d deposits. Humans with aceruloplasminemia causing RPE iron overload had increased RPE C3d deposition. The molecular events in the iron-C3 pathway represent therapeutic targets for AMD or other diseases exacerbated by iron-induced local complement dysregulation.
Collapse
Affiliation(s)
- Yafeng Li
- From the F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute
| | - Delu Song
- From the F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute
| | - Ying Song
- From the F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute
| | - Liangliang Zhao
- From the F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute
| | - Natalie Wolkow
- From the F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute
| | | | - Wenchao Song
- Department of Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Joshua L Dunaief
- From the F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute,
| |
Collapse
|
41
|
Reichl P, Dengler M, van Zijl F, Huber H, Führlinger G, Reichel C, Sieghart W, Peck-Radosavljevic M, Grubinger M, Mikulits W. Axl activates autocrine transforming growth factor-β signaling in hepatocellular carcinoma. Hepatology 2015; 61:930-41. [PMID: 25251599 PMCID: PMC4450343 DOI: 10.1002/hep.27492] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022]
Abstract
UNLABELLED In hepatocellular carcinoma (HCC), intrahepatic metastasis frequently correlates with epithelial to mesenchymal transition (EMT) of malignant hepatocytes. Several mechanisms have been identified to be essentially involved in hepatocellular EMT, among them transforming growth factor (TGF)-β signaling. Here we show the up-regulation and activation of the receptor tyrosine kinase Axl in EMT-transformed hepatoma cells. Knockdown of Axl expression resulted in abrogation of invasive and transendothelial migratory abilities of mesenchymal HCC cells in vitro and Axl overexpression-induced metastatic colonization of epithelial hepatoma cells in vivo. Importantly, Axl knockdown severely impaired resistance to TGF-β-mediated growth inhibition. Analysis of the Axl interactome revealed binding of Axl to 14-3-3ζ, which is essentially required for Axl-mediated cell invasion, transendothelial migration, and resistance against TGF-β. Axl/14-3-3ζ signaling caused phosphorylation of Smad3 linker region (Smad3L) at Ser213, resulting in the up-regulation of tumor-progressive TGF-β target genes such as PAI1, MMP9, and Snail as well as augmented TGF-β1 secretion in mesenchymal HCC cells. Accordingly, high Axl expression in HCC patient samples correlated with elevated vessel invasion of HCC cells, higher risk of tumor recurrence after liver transplantation, strong phosphorylation of Smad3L, and lower survival. In addition, elevated expression of both Axl and 14-3-3ζ showed strongly reduced survival of HCC patients. CONCLUSION Our data suggest that Axl/14-3-3ζ signaling is central for TGF-β-mediated HCC progression and a promising target for HCC therapy.
Collapse
Affiliation(s)
- Patrick Reichl
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Mirko Dengler
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Franziska van Zijl
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Heidemarie Huber
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gerhard Führlinger
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Wolfgang Sieghart
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Markus Peck-Radosavljevic
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Markus Grubinger
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Phosphorylation of Smad2/3 at specific linker threonine indicates slow-cycling intestinal stem-like cells before reentry to cell cycle. Dig Dis Sci 2015; 60:362-74. [PMID: 25185661 DOI: 10.1007/s10620-014-3348-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 08/25/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Quiescent (slow-cycling) and active (rapid-cycling) stem cells are demonstrated in small intestines. We have identified significant expression of Smad2/3, phosphorylated at specific linker threonine residues (pSmad2/3L-Thr), in murine stomach, and suggested these cells are epithelial stem cells. AIM Here, we explore whether pSmad2/3L-Thr could serve as a biomarker for small intestine and colon stem cells. METHODS We examined small intestines and colons from C57BL/6 mice and colons with dextran sulfate sodium (DSS)-induced colitis. We performed double-immunofluorescent staining of pSmad2/3L-Thr with Ki67, cytokeratin 8, chromogranin A, CDK4, DCAMKL1, and Musashi-1. Small intestines and colons from Lgr5-EGFP knock-in mice were examined by pSmad2/3L-Thr immunofluorescent staining. To examine BrdU label retention of pSmad2/3L-Thr immunostaining-positive cells, we collected specimens after BrdU administration and observed double-immunofluorescent staining of pSmad2/3L-Thr with BrdU. RESULTS In small intestines and colons, pSmad2/3L-Thr immunostaining-strongly positive cells were detected around crypt bases. Immunohistochemical co-localization of pSmad2/3L-Thr with Ki67 was not observed. pSmad2/3L-Thr immunostaining-strongly positive cells showed co-localization with cytokeratin 8, CDK4, and Musashi-1 and different localization from chromogranin A and DCAMKL1 immunostaining-positive cells. Under a light microscope, pSmad2/3L-Thr immunostaining-strongly positive cells were morphologically undifferentiated. In Lgr5-EGFP knock-in mice, some but not all pSmad2/3L-Thr immunostaining-strongly positive cells showed co-localization with Lgr5. pSmad2/3L-Thr immunostaining-strongly positive cells showed co-localization with BrdU at 5, 10, and 15 days after administration. In DSS-induced colitis, pSmad2/3L-Thr and Ki67 immunostaining-positive cells increased in the regeneration phase and decreased in the injury phase. CONCLUSION In murine small intestines and colons, we suggest pSmad2/3L-Thr immunostaining-strongly positive cells are epithelial stem-like cells just before reentry to the cell cycle.
Collapse
|
43
|
Zhang L, Liu C, Meng XM, Huang C, Xu F, Li J. Smad2 protects against TGF-β1/Smad3-mediated collagen synthesis in human hepatic stellate cells during hepatic fibrosis. Mol Cell Biochem 2014; 400:17-28. [PMID: 25351340 DOI: 10.1007/s11010-014-2258-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 10/17/2014] [Indexed: 12/11/2022]
Abstract
With structural similarity but functional diversity, Smad2 and Smad3 interact with each other to mediate transforming growth factor-β (TGF-β)-triggered signaling transduction. However, in the hepatic fibrosis, the detailed roles of R-Smads, and interaction between Smad2 and Smad3 are still undefined. In this setting, we established a rat model of CCl4-induced hepatic fibrosis in vivo and TGF-β1-treated hepatic stellate cell model in vitro to detect whether Smad2 and Smad3 play distinct roles in mediating liver fibrogenesis. Results indicated that both phosphorylation of Smad2 and Smad3 were detected in the hepatic stellate cells of liver fibrotic tissues and cells. Furthermore, In vitro data demonstrated that knockdown of Smad2 in human hepatic stellate cells increased expression of collagen I (Col.I), tissue inhibitor of metalloproteinase-1 (TIMP-1) whereas decreasing expression of the matrix metalloproteinases-2(MMP-2) in presence of TGF-β1 compared with control group. In contrast, knockdown of Smad3 significantly reduced TGF-β1-induced Col.I production. These findings were further evident by the results that overexpression of Smad2 attenuated the expression of Col.I and TIMP-1, but enhanced MMP-2 whereas overexpression of Smad3 showed the opposite effect. Furthermore, Smad2 suppressed the phosphorylation and nuclear translocation of Smad3, which may protect against Smad3-mediated fibrotic response. Collectively, Smad2 may be a potential therapeutic target for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | | | | | | | | | | |
Collapse
|
44
|
Fang J, Xu H, Yang C, Morsalin S, Kayarthodi S, Rungsrisuriyachai K, Gunnal U, Mckenzie B, Rao VN, Reddy ESP. Ets Related Gene and Smad3 Proteins Collaborate to Activate Transforming Growth Factor-Beta Mediated Signaling Pathway in ETS Related Gene-Positive Prostate Cancer Cells. ACTA ACUST UNITED AC 2014; 1:175-181. [PMID: 25745638 DOI: 10.1166/jpsp.2014.1022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
TGF-β/Smads signaling plays a significant role in the regulation of growth of normal and prostate cancer cells. Smad proteins function as important mediators of intracellular signal transduction of transforming growth factor-β (TGF-β). TGF-β signaling pathway is known to regulate cell proliferation, differentiation, apoptosis and play a major role in some human diseases and cancers. Following their phosphorylation by TGF-β receptor-I, Receptor-regulated Smads (including Smad2 and Smad3 proteins) form a heteromeric complex with co-Smad (Smad4) and then translocate into the nucleus where they bind and regulate the expression of target genes. ERG (Ets Related Gene) belongs to the ETS family of transcriptional factors. Chromosomal rearrangement of TMPRSS2 gene and ERG gene has been found in majority of prostate cancers. Over-expression of full length or truncated ERG proteins have been shown to associate with a higher rate of recurrent and unfavorable prognosis of prostate cancer. In order to understand how ERG oncoprotein regulates TGF-β/Smads signaling pathway, we have studied the effect of ERG on TGF-β/Smad3 signaling pathway. In this study, we demonstrate that ERG oncoprotein physically interacts with Smad3 protein and stabilizes phospho-Smad3 protein and thereby enhance TGF-β/Smad3 signaling pathway in prostate cells. Thus, ERG oncoprotein plays an important role in prostate tumorigenesis by using a novel mechanism to activate TGF-β/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Jinbo Fang
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Huali Xu
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Chunshu Yang
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Sharif Morsalin
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Shubhalaxmi Kayarthodi
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Kunchala Rungsrisuriyachai
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Ujwala Gunnal
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA ; Department of Medicine, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Brittany Mckenzie
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - Veena N Rao
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| | - E Shyam P Reddy
- Cancer Biology Program, Department of OB/GYN, Morehouse School of Medicine, Georgia Cancer Center for Excellence, Grady Memorial Hospital, 80 Jesse Hill Jr. Drive, Atlanta, GA 30303, USA
| |
Collapse
|
45
|
McCubrey JA, Davis NM, Abrams SL, Montalto G, Cervello M, Libra M, Nicoletti F, D'Assoro AB, Cocco L, Martelli AM, Steelman LS. Targeting breast cancer initiating cells: advances in breast cancer research and therapy. Adv Biol Regul 2014; 56:81-107. [PMID: 24913694 DOI: 10.1016/j.jbior.2014.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/06/2014] [Indexed: 06/03/2023]
Abstract
Over the past 10 years there have been significant advances in our understanding of breast cancer and the important roles that breast cancer initiating cells (CICs) play in the development and resistance of breast cancer. Breast CICs endowed with self-renewing and tumor-initiating capacities are believed to be responsible for the relapses which often occur after various breast cancer therapies. In this review, we will summarize some of the key developments in breast CICs which will include discussion of some of the key genes implicated: estrogen receptor (ER), HER2, BRCA1, TP53, PIK3CA, RB, P16INK1 and various miRs as well some drugs which are showing promise in targeting CICs. In addition, the concept of combined therapies will be discussed. Basic and clinical research is resulting in novel approaches to improve breast cancer therapy by targeting the breast CICs.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA.
| | - Nicole M Davis
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-Medical Sciences, University of Catania, Catania, Italy
| | | | - Antonino B D'Assoro
- Department of Medical Oncology, Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA
| |
Collapse
|
46
|
Takahashi Y, Fukui T, Kishimoto M, Suzuki R, Mitsuyama T, Sumimoto K, Okazaki T, Sakao M, Sakaguchi Y, Yoshida K, Uchida K, Nishio A, Matsuzaki K, Okazaki K. Phosphorylation of Smad2/3 at the specific linker threonine residue indicates slow-cycling esophageal stem-like cells before re-entry to the cell cycle. Dis Esophagus 2014; 29:107-15. [PMID: 25168378 DOI: 10.1111/dote.12277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED The stem cell compartment in the esophageal epithelium is possibly located in the basal layer. We have identified significant expression of Smad2/3, phosphorylated at specific linker threonine residues (pSmad2/3L-Thr), in the epithelial cells of murine stomach and intestine, and have suggested that these cells are epithelial stem cells. In this study, we explore whether pSmad2/3L-Thr could serve as a biomarker for esophageal stem cells. We examined esophageal tissues from normal C57BL/6 mice and those with esophagitis. Double immunofluorescent staining of pSmad2/3L-Thr with Ki67, CDK4, p63, or CK14 was performed. After immunofluorescent staining, we stained the same sections with hematoxylin-eosin and observed these cells under a light microscope. We used the 5-bromo-2-deoxyuridine (BrdU) labeling assay to examine label retention of pSmad2/3L-Thr immunostaining-positive cells. We collected specimens 5, 10, 15 and 20 days after repeated BrdU administrations and observed double immunofluorescent staining of pSmad2/3L-Thr with BrdU. In the esophagus, pSmad2/3L-Thr immunostaining-positive cells were detected in the basal layer. These cells were detected between Ki67 immunostaining-positive cells, but they were not co-localized with Ki67. pSmad2/3L-Thr immunostaining-positive cells showed co-localization with CDK4, p63, and CK14. Under a light microscope, pSmad2/3L-Thr immunostaining-positive cells indicated undifferentiated morphological features. Until 20 days follow-up period, pSmad2/3L-Thr immunostaining-positive cells were co-localized with BrdU. pSmad2/3L-Thr immunostaining-positive cells significantly increased in the regeneration phase of esophagitis mucosae, as compared with control mice (esophagitis vs. CONTROL 6.889 ± 0.676/cm vs. 4.293 ± 0.659/cm; P < 0.001). We have identified significant expression of pSmad2/3L-Thr in the specific epithelial cells of murine esophagi. We suggest that these cells are slow-cycling epithelial stem-like cells before re-entry to the cell cycle.
Collapse
Affiliation(s)
- Y Takahashi
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - T Fukui
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - M Kishimoto
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - R Suzuki
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - T Mitsuyama
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - K Sumimoto
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - T Okazaki
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - M Sakao
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - Y Sakaguchi
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - K Yoshida
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - K Uchida
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - A Nishio
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - K Matsuzaki
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| | - K Okazaki
- The Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
47
|
Tarasewicz E, Hamdan R, Straehla J, Hardy A, Nunez O, Zelivianski S, Dokic D, Jeruss JS. CDK4 inhibition and doxorubicin mediate breast cancer cell apoptosis through Smad3 and survivin. Cancer Biol Ther 2014; 15:1301-11. [PMID: 25006666 DOI: 10.4161/cbt.29693] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cyclin D1/CDK4 activity is upregulated in up to 50% of breast cancers and CDK4-mediated phosphorylation negatively regulates the TGFβ superfamily member Smad3. We sought to determine if CDK4 inhibition and doxorubicin chemotherapy could impact Smad3-mediated cell/colony growth and apoptosis in breast cancer cells. Parental and cyclin D1-overexpressing MCF7 cells were treated with CDK4 inhibitor, doxorubicin, or combination therapy and cell proliferation, apoptosis, colony formation, and expression of apoptotic proteins were evaluated using an MTS assay, TUNEL staining, 3D Matrigel assay, and apoptosis array/immunoblotting. Study cells were also transduced with WT Smad3 or a Smad3 construct resistant to CDK4 phosphorylation (5M) and colony formation and expression of apoptotic proteins were assessed. Treatment with CDK4 inhibitor/doxorubicin combination therapy, or transduction with 5M Smad3, resulted in a similar decrease in colony formation. Treating cyclin D overexpressing breast cancer cells with combination therapy also resulted in the greatest increase in apoptosis, resulted in decreased expression of anti-apoptotic proteins survivin and XIAP, and impacted subcellular localization of pro-apoptotic Smac/DIABLO. Additionally, transduction of 5M Smad3 and doxorubicin treatment resulted in the greatest change in apoptotic protein expression. Collectively, this work showed the impact of CDK4 inhibitor-mediated, Smad3-regulated tumor suppression, which was augmented in doxorubicin-treated cyclin D-overexpressing study cells.
Collapse
Affiliation(s)
- Elizabeth Tarasewicz
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Randala Hamdan
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Joelle Straehla
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Ashley Hardy
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Omar Nunez
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Stanislav Zelivianski
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Danijela Dokic
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| | - Jacqueline S Jeruss
- Department of Surgery; Northwestern University Feinberg School of Medicine; Chicago, IL USA; Robert H. Lurie Comprehensive Cancer Center; Chicago, IL USA
| |
Collapse
|
48
|
Abstract
The circadian clock is an endogenous timekeeper system that controls the daily rhythms of a variety of physiological processes. Accumulating evidence indicates that genetic changes or unhealthy lifestyle can lead to a disruption of circadian homeostasis, which is a risk factor for severe dysfunctions and pathologies including cancer. Cell cycle, proliferation, and cell death are closely intertwined with the circadian clock, and thus disruption of circadian rhythms appears to be linked to cancer development and progression. At the molecular level, the cell cycle machinery and the circadian clocks are controlled by similar mechanisms, including feedback loops of genes and protein products that display periodic activation and repression. Here, we review the circadian rhythmicity of genes associated with the cell cycle, proliferation, and apoptosis, and we highlight the potential connection between these processes, the circadian clock, and neoplastic transformations. Understanding these interconnections might have potential implications for the prevention and therapy of malignant diseases.
Collapse
Affiliation(s)
- Matúš Soták
- Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | | | | |
Collapse
|
49
|
Zhang Z, Wu L, Wang J, Li G, Feng D, Zhang B, Li L, Yang J, Ma L, Qin H. Opposing effects of PI3K/Akt and Smad-dependent signaling pathways in NAG-1-induced glioblastoma cell apoptosis. PLoS One 2014; 9:e96283. [PMID: 24759784 PMCID: PMC3997521 DOI: 10.1371/journal.pone.0096283] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 04/05/2014] [Indexed: 11/18/2022] Open
Abstract
Nonsteroidal anti-inflammatory drug (NSAID) activated gene-1 (NAG-1) is a divergent member of the transforming growth factor-beta (TGF-β) superfamily. NAG-1 plays remarkable multifunctional roles in controlling diverse physiological and pathological processes including cancer. Like other TGF-β family members, NAG-1 can play dual roles during cancer development and progression by negatively or positively modulating cancer cell behaviors. In glioblastoma brain tumors, NAG-1 appears to act as a tumor suppressor gene; however, the precise underlying mechanisms have not been well elucidated. In the present study, we discovered that overexpression of NAG-1 induced apoptosis in U87 MG, U118 MG, U251 MG, and T98G cell lines via the intrinsic mitochondrial pathway, but not in A172 and LN-229 cell lines. NAG-1 could induce the phosphorylation of PI3K/Akt and Smad2/3 in all six tested glioblastoma cell lines, except Smad3 phosphorylation in A172 and LN-229 cell lines. In fact, Smad3 expression and its phosphorylation were almost undetectable in A172 and LN-229 cells. The PI3K inhibitors promoted NAG-1-induced glioblastoma cell apoptosis, while siRNAs to Smad2 and Smad3 decreased the apoptosis rate. NAG-1 also stimulated the direct interaction between Akt and Smad3 in glioblastoma cells. Elevating the level of Smad3 restored the sensitivity to NAG-1-induced apoptosis in A172 and LN-229 cells. In conclusion, our results suggest that PI3K/Akt and Smad-dependent signaling pathways display opposing effects in NAG-1-induced glioblastoma cell apoptosis.
Collapse
Affiliation(s)
- Zhiguo Zhang
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- Postdoctoral research station of Neurosurgery, Wuhan General Hospital of Guangzhou Command, PLA, Wuhan, China
| | - Lin Wu
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi’an, China
| | - Julei Wang
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Gang Li
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Dayun Feng
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Bin Zhang
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Lihong Li
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jiandong Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Lianting Ma
- Postdoctoral research station of Neurosurgery, Wuhan General Hospital of Guangzhou Command, PLA, Wuhan, China
| | - Huaizhou Qin
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- * E-mail:
| |
Collapse
|
50
|
Lee IH, Sohn M, Lim HJ, Yoon S, Oh H, Shin S, Shin JH, Oh SH, Kim J, Lee DK, Noh DY, Bae DS, Seong JK, Bae YS. Ahnak functions as a tumor suppressor via modulation of TGFβ/Smad signaling pathway. Oncogene 2014; 33:4675-84. [PMID: 24662814 PMCID: PMC4180639 DOI: 10.1038/onc.2014.69] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 12/15/2013] [Accepted: 12/24/2013] [Indexed: 02/06/2023]
Abstract
We provide detailed mechanisms of Ahnak-mediated potentiation of transforming growth factor β (TGFβ) signaling, which leads to a negative regulation of cell growth. We show that Smad3 interacts with Ahnak through MH2 domain and that Ahnak stimulates Smad3 localization into nucleus leading to potentiating TGFβ-induced transcriptional activity of R-Smad. Moreover, overexpression of Ahnak resulted in growth retardation and cell cycle arrest through downregulation of c-Myc and cyclin D1/D2. We describe results from analyses of Ahnak−/− mouse model expressing middle T antigen in a mammary gland-specific manner (MMTVTg/+Ahnak−/−), which showed significantly progressed hyperplasia of mammary glands compared with MMTVTg/+Ahnak+/+. Finally, we screened multiple human breast cancer tissues and showed that the expression of Ahnak in cancer tissues is lower than that in control tissues by 50%. Taken together, these data indicate that Ahnak mediates a negative regulation of cell growth and acts as novel tumor suppressor through potentiation of TGFβ signaling.
Collapse
Affiliation(s)
- I H Lee
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - M Sohn
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - H J Lim
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - S Yoon
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - H Oh
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - S Shin
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - J H Shin
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - S-H Oh
- College of Pharmacy, Gachon University, Incheon, Korea
| | - J Kim
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - D K Lee
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| | - D Y Noh
- Department of Surgery, School of Medicine, Seoul National University, Seoul, Korea
| | - D S Bae
- Department of Obstetrics and Gynecology, Samsung Hospital, SungKyunKwan University, Seoul, Korea
| | - J K Seong
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Y S Bae
- Department of Life Sciences and GT5 program, Ewha Womans University, Seoul, Korea
| |
Collapse
|