51
|
Mehdipour M, Liu Y, Liu C, Kumar B, Kim D, Gathwala R, Conboy IM. Key Age-Imposed Signaling Changes That Are Responsible for the Decline of Stem Cell Function. Subcell Biochem 2018; 90:119-143. [PMID: 30779008 DOI: 10.1007/978-981-13-2835-0_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This chapter analyzes recent developments in the field of signal transduction of ageing with the focus on the age-imposed changes in TGF-beta/pSmad, Notch, Wnt/beta-catenin, and Jak/Stat networks. Specifically, this chapter delineates how the above-mentioned evolutionary-conserved morphogenic signaling pathways operate in young versus aged mammalian tissues, with insights into how the age-specific broad decline of stem cell function is precipitated by the deregulation of these key cell signaling networks. This chapter also provides perspectives onto the development of defined therapeutic approaches that aim to calibrate intensity of the determinant signal transduction to health-youth, thereby rejuvenating multiple tissues in older people.
Collapse
Affiliation(s)
- Melod Mehdipour
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA
| | - Yutong Liu
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA
| | - Chao Liu
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA
| | - Binod Kumar
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA
| | - Daehwan Kim
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA
| | - Ranveer Gathwala
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA
| | - Irina M Conboy
- Bioengineering, Univercity of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
52
|
Li L, Liu CC, Chen X, Xu S, Hernandez Cortes-Manno S, Cheng SH. Mechanistic Study of Bakuchiol-Induced Anti-breast Cancer Stem Cell and in Vivo Anti-metastasis Effects. Front Pharmacol 2017; 8:746. [PMID: 29093680 PMCID: PMC5651275 DOI: 10.3389/fphar.2017.00746] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022] Open
Abstract
Cancer stem cells are involved in cancer establishment, progression, and resistance to current treatments. We demonstrated the in vitro and in vivo anti-breast cancer effect of bakuchiol in a previous study. However, the ability of bakuchiol to target breast cancer stem cells (BCSCs) and inhibit breast cancer metastasis remains unknown. In the current study, we used the cell surface markers CD44 and CD24 to distinguish BCSCs from MCF-7 cells. Bakuchiol inhibited mammosphere formation and aldehyde dehydrogenase activity in BCSCs. Moreover, bakuchiol induced apoptosis and suppressed the mitochondrial membrane potential of BCSCs. Bakuchiol upregulated the expression levels of pro-apoptotic genes, BNIP3 and DAPK2. Bakuchiol induced oxidative stress and altered lipogenesis in BCSCs. In zebrafish xenografts, bakuchiol inhibited breast cancer cell metastasis in vivo. In addition, bakuchiol altered the expression levels of metastasis-related genes through upregulating CK18 and downregulating Notch3, FASN, TGFBR1, and ACVR1B. Our study provides evidence for the anti-breast cancer potential of bakuchiol.
Collapse
Affiliation(s)
- Li Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Chi C Liu
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Xueping Chen
- Vitargent (International) Biotechnology Limited, Sha Tin, Hong Kong
| | - Shisan Xu
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Kowloon, Hong Kong
| | | | - Shuk H Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
53
|
Villalba M, Evans SR, Vidal-Vanaclocha F, Calvo A. Role of TGF-β in metastatic colon cancer: it is finally time for targeted therapy. Cell Tissue Res 2017; 370:29-39. [DOI: 10.1007/s00441-017-2633-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/24/2017] [Indexed: 12/15/2022]
|
54
|
Yuan Y, Yang M, Wang K, Sun J, Song L, Diao X, Jiang Z, Cheng G, Wang X. Excessive activation of the TLR9/TGF-β1/PDGF-B pathway in the peripheral blood of patients with systemic lupus erythematosus. Arthritis Res Ther 2017; 19:70. [PMID: 28356164 PMCID: PMC5372299 DOI: 10.1186/s13075-017-1238-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/16/2017] [Indexed: 11/13/2022] Open
Abstract
Background Our aim is to study the existence of the TLR9/TGF-β1/PDGF-B pathway in healthy humans and patients with systemic lupus erythematosus (SLE), and to explore its possible involvement in the pathogenesis of lupus nephritis (LN). Methods Protein levels of the cytokines were detected by ELISA. mRNA levels of the cytokines were analyzed by real-time PCR. MTT assay was used to test the proliferation of mesangial cells under different treatments. Results Compared to healthy controls (NControl = 56), levels of Toll-like receptor (TLR)9, transforming growth factor (TGF)-β1, and platelet-derived growth factor B (PDGF-B) were increased significantly in the peripheral blood of SLE patients (NSLE = 112). Significant correlations between the levels of TLR9, TGF-β1, and PDGF-B were observed in both healthy controls and SLE patients. The levels of TGF-β1 and PDGF-B were greatly enhanced by TLR9 activation in primary cell cultures. The proliferation of mesangial cells induced by the plasma of SLE patients was significantly higher than that induced by healthy controls; PDGF-B was involved in this process. The protein levels of PDGF-B homodimer correlated with the levels of urine protein in SLE patients with LN (NLN =38). Conclusions The TLR9/TGF-β1/PDGF-B pathway exists in humans and can be excessively activated in SLE patients. High levels of PDGF-B may result in overproliferation of mesangial cells in the kidney that are involved in the development of glomerulonephritis and LN. Further studies are necessary to identify TLR9, TGF-β1, and PDGF-B as new therapeutic targets to prevent the development of glomerulonephritis and LN. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1238-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yi Yuan
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China.,Department of Rheumatology and Immunology, the First Hospital, Jilin University, Changchun, 130021, China
| | - Mingyue Yang
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China
| | - Kuo Wang
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China
| | - Jing Sun
- Shanghai Wisdom Chemical Research Co. Ltd., Shanghai, 201203, China
| | - Lili Song
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China
| | - Xue Diao
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China
| | - Zhenyu Jiang
- Department of Rheumatology and Immunology, the First Hospital, Jilin University, Changchun, 130021, China.
| | - Genhong Cheng
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China. .,Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Xiaosong Wang
- Institute of Translational Medicine, the First Hospital, Jilin University, Changchun, 130061, China.
| |
Collapse
|
55
|
Sun N, Taguchi A, Hanash S. Switching Roles of TGF-β in Cancer Development: Implications for Therapeutic Target and Biomarker Studies. J Clin Med 2016; 5:jcm5120109. [PMID: 27916872 PMCID: PMC5184782 DOI: 10.3390/jcm5120109] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 12/15/2022] Open
Abstract
TGF-β induces complicated and even opposite responses in numerous biological processes, e.g., tumor suppression in pre-malignant cells and metastasis promotion in cancer cells. However, the cellular contextual determinants of these different TGF-β roles remain elusive, and the driver genes triggering the determinants’ changes have not been identified. Recently, however, several findings have provided new insights on the contextual determinants of Smads in TGF-β’s biological processes. These novel switches and their effectors may serve as prognostic biomarkers and therapeutic targets of TGF-β-mediated cancer progression.
Collapse
Affiliation(s)
- Nan Sun
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Ayumu Taguchi
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Samir Hanash
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
56
|
Kim EJ, Kang JI, Tung NH, Kim YH, Hyun JW, Koh YS, Chang WY, Yoo ES, Kang HK. The Effect of (1S,2S,3E,7E,11E)-3,7,11,15-Cembratetraen-17,2-Olide (LS-1) from Lobophyyum sp. on the Apoptosis Induction of SNU-C5 Human Colorectal Cancer Cells. Biomol Ther (Seoul) 2016; 24:623-629. [PMID: 27469141 PMCID: PMC5098542 DOI: 10.4062/biomolther.2016.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/04/2016] [Accepted: 05/12/2016] [Indexed: 01/07/2023] Open
Abstract
(1S,2S,3E,7E,11E)-3,7,11,15-cembratetraen-17,2-olide (LS-1), a marine cembrenolide diterpene, has anticancer activity against colon cancer cells such as HT-29, SNU-C5/5-FU (fluorouracil-resistant SNU-C5) and SNU-C5. However, the action mechanism of LS-1 on SNU-C5 human colon cancer cells has not been fully elucidated. In this study, we investigated whether the anticancer effect of LS-1 could result from apoptosis via the modulation of Wnt/β-catenin and the TGF-β pathways. When treated with the LS-1, we could observe the apoptotic characteristics such as apoptotic bodies and the increase of sub-G1 hypodiploid cell population, increase of Bax level, decrease of Bcl-2 expression, cleavage of procaspase-3 and cleavage of poly (ADP-ribose) polymerase in SNU-C5 cells. Furthermore, the apoptosis induction of SNU-C5 cells upon LS-1 treatment was also accompanied by the down-regulation of Wnt/β-catenin signaling pathway via the decrease of GSK-3β phosphorylation followed by the decrease of β-catenin level. In addition, the LS-1 induced the activation of TGF-β signaling pathway with the decrease of carcinoembryonic antigen which leads to decrease of c-Myc, an oncoprotein. These data suggest that the LS-1 could induce the apoptosis via the down-regulation of Wnt/β-catenin pathway and the activation of TGF-β pathway in SNU-C5 human colon cancer cells. The results support that the LS-1 might have potential for the treatment of human colon cancer.
Collapse
Affiliation(s)
- Eun-Ji Kim
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jung Il Kang
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Nguyen-Huu Tung
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin Won Hyun
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Young Sang Koh
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Weon-Young Chang
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Eun Sook Yoo
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Hee-Kyoung Kang
- School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
57
|
Ivanović V, Dedović-Tanić N, Milovanović Z, Lukić S, Nikolić S, Baltić V, Stojiljković B, Demajo M, Mandušić V, Dimitrijević B. Case with triple-negative breast cancer shows overexpression of both cFOS and TGF-β1 in node-positive tissue. Per Med 2016; 13:523-530. [PMID: 29754549 DOI: 10.2217/pme-2016-0032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We present herein a case report style article on a rare advanced triple-negative breast cancer (TNBC) patient with 6-month disease-free interval, and 10-month overall survival. Our results demonstrate that the poor clinical outcome of this patient was associated with pronounced, more than fivefold higher, overexpression of both cFOS and TGF-β1 proteins in its metastatic nodal tissue extracts, when compared with the values of the two non-TNBC controls (with 'zero' disease-free interval and overall survival). This original observation suggests, for the first time, that both the cFOS and TGF-β1 may be considered as a pair of biomarkers for an early assessment of poor prognosis for TNBC patients. The possible clinical implication of this observation is discussed.
Collapse
Affiliation(s)
- Vesna Ivanović
- Department for Radiobiology & Molecular Genetics, Institute of Nuclear Sciences 'Vinča', University of Belgrade, Belgrade, Serbia
| | - Nasta Dedović-Tanić
- Department for Radiobiology & Molecular Genetics, Institute of Nuclear Sciences 'Vinča', University of Belgrade, Belgrade, Serbia
| | | | - Silvana Lukić
- Institute of Oncology & Radiology of Serbia, Belgrade, Serbia
| | - Srdjan Nikolić
- Institute of Oncology & Radiology of Serbia, Belgrade, Serbia
| | | | | | - Miroslav Demajo
- Department for Radiobiology & Molecular Genetics, Institute of Nuclear Sciences 'Vinča', University of Belgrade, Belgrade, Serbia
| | - Vesna Mandušić
- Department for Radiobiology & Molecular Genetics, Institute of Nuclear Sciences 'Vinča', University of Belgrade, Belgrade, Serbia
| | - Bogomir Dimitrijević
- Department for Radiobiology & Molecular Genetics, Institute of Nuclear Sciences 'Vinča', University of Belgrade, Belgrade, Serbia
| |
Collapse
|
58
|
Candia J. TGF-ß alterations in oral squamous cell carcinoma. Narrative review. JOURNAL OF ORAL RESEARCH 2016. [DOI: 10.17126/joralres.2016.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
59
|
Ng S, Deng J, Chinnadurai R, Yuan S, Pennati A, Galipeau J. Stimulation of Natural Killer Cell-Mediated Tumor Immunity by an IL15/TGFβ-Neutralizing Fusion Protein. Cancer Res 2016; 76:5683-5695. [PMID: 27488533 DOI: 10.1158/0008-5472.can-16-0386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/19/2016] [Indexed: 11/16/2022]
Abstract
The clinical efficacy of immune cytokines used for cancer therapy is hampered by elements of the immunosuppressive tumor microenvironment such as TGFβ. Here we demonstrate that FIST15, a recombinant chimeric protein composed of the T-cell-stimulatory cytokine IL15, the sushi domain of IL15Rα and a TGFβ ligand trap, can overcome immunosuppressive TGFβ to effectively stimulate the proliferation and activation of natural killer (NK) and CD8+ T cells with potent antitumor properties. FIST15-treated NK and CD8+ T cells produced more IFNγ and TNFα compared with treatment with IL15 and a commercially available TGFβ receptor-Fc fusion protein (sTβRII) in the presence of TGFβ. Murine B16 melanoma cells, which overproduce TGFβ, were lysed by FIST15-treated NK cells in vitro at doses approximately 10-fold lower than NK cells treated with IL15 and sTβRII. Melanoma cells transduced to express FIST15 failed to establish tumors in vivo in immunocompetent murine hosts and could only form tumors in beige mice lacking NK cells. Mice injected with the same cells were also protected from subsequent challenge by unmodified B16 melanoma cells. Finally, mice with pre-established B16 melanoma tumors responded to FIST15 treatment more strongly compared with tumors treated with control cytokines. Taken together, our results offer a preclinical proof of concept for the use of FIST15 as a new class of biological therapeutics that can coordinately neutralize the effects of immunosuppressive TGFβ in the tumor microenvironment while empowering tumor immunity. Cancer Res; 76(19); 5683-95. ©2016 AACR.
Collapse
Affiliation(s)
- Spencer Ng
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jiusheng Deng
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Raghavan Chinnadurai
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Shala Yuan
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Andrea Pennati
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jacques Galipeau
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia. Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia.
| |
Collapse
|
60
|
Xiao H, Zhang J, Xu Z, Feng Y, Zhang M, Liu J, Chen R, Shen J, Wu J, Lu Z, Fang X, Li J, Zhang Y. Metformin is a novel suppressor for transforming growth factor (TGF)-β1. Sci Rep 2016; 6:28597. [PMID: 27349853 PMCID: PMC4923858 DOI: 10.1038/srep28597] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/07/2016] [Indexed: 11/09/2022] Open
Abstract
Metformin is a widely used first-line antidiabetic drug that has been shown to protect against a variety of specific diseases in addition to diabetes, including cardiovascular disorders, polycystic ovary syndrome, and cancer. However, the precise mechanisms underlying the diverse therapeutic effects of metformin remain elusive. Here, we report that transforming growth factor-β1 (TGF-β1), which is involved in the pathogenesis of numerous diseases, is a novel target of metformin. Using a surface plasmon resonance-based assay, we identified the direct binding of metformin to TGF-β1 and found that metformin inhibits [125I]-TGF-β1 binding to its receptor. Furthermore, based on molecular docking and molecular dynamics simulations, metformin was predicted to interact with TGF-β1 at its receptor-binding domain. Single-molecule force spectroscopy revealed that metformin reduces the binding probability but not the binding force of TGF-β1 to its type II receptor. Consequently, metformin suppresses type II TGF-β1 receptor dimerization upon exposure to TGF-β1, which is essential for downstream signal transduction. Thus, our results indicate that metformin is a novel TGF-β suppressor with therapeutic potential for numerous diseases in which TGF-β1 hyperfunction is indicated.
Collapse
Affiliation(s)
- Han Xiao
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jianshu Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Zhonghe Xu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Yenan Feng
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Mingliang Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jianli Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructures and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ruifei Chen
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jing Shen
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jimin Wu
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Zhizhen Lu
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Xiaohong Fang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructures and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jingyuan Li
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
61
|
Carotta S. Targeting NK Cells for Anticancer Immunotherapy: Clinical and Preclinical Approaches. Front Immunol 2016; 7:152. [PMID: 27148271 PMCID: PMC4838611 DOI: 10.3389/fimmu.2016.00152] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/07/2016] [Indexed: 11/23/2022] Open
Abstract
The recent success of checkpoint blockade has highlighted the potential of immunotherapy approaches for cancer treatment. Although the majority of approved immunotherapy drugs target T cell subsets, it is appreciated that other components of the immune system have important roles in tumor immune surveillance as well and thus represent promising additional targets for immunotherapy. Natural killer (NK) cells are the body’s first line of defense against infected or transformed cells, as they kill target cells in an antigen-independent manner. Although several studies have clearly demonstrated the active role of NK cells in cancer immune surveillance, only few clinically approved therapies currently exist that harness their potential. Our increased understanding of NK cell biology over the past few years has renewed the interest in NK cell-based anticancer therapies, which has lead to a steady increase of NK cell-based clinical and preclinical trials. Here, the role of NK cells in cancer immune surveillance is summarized, and several novel approaches to enhance NK cell cytotoxicity against cancer are discussed.
Collapse
Affiliation(s)
- Sebastian Carotta
- Immune Modulation Department, Boehringer Ingelheim RCV, Vienna, Austria; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| |
Collapse
|
62
|
Charbonneau M, Lavoie RR, Lauzier A, Harper K, McDonald PP, Dubois CM. Platelet-Derived Growth Factor Receptor Activation Promotes the Prodestructive Invadosome-Forming Phenotype of Synoviocytes from Patients with Rheumatoid Arthritis. THE JOURNAL OF IMMUNOLOGY 2016; 196:3264-75. [DOI: 10.4049/jimmunol.1500502] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 02/15/2016] [Indexed: 11/19/2022]
|
63
|
Jaschinski F, Korhonen H, Janicot M. Design and Selection of Antisense Oligonucleotides Targeting Transforming Growth Factor Beta (TGF-β) Isoform mRNAs for the Treatment of Solid Tumors. Methods Mol Biol 2016; 1317:137-51. [PMID: 26072406 DOI: 10.1007/978-1-4939-2727-2_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Transforming growth factor beta isoforms (TGF-β1, -β2, and -β3) are cytokines associated with a wide range of biological processes in oncology including tumor cell invasion and migration, angiogenesis, immunosuppression, as well as regulation of tumor stem cell properties. Hence, blocking the TGF-β signaling pathways may have a multifold therapeutic benefit for the treatment of solid tumors. Here, we describe the identification and selection processes for the development of highly potent and selective chemically modified antisense oligodeoxynucleotides (fully phosphorothioate locked nucleic acid gapmers) allowing effective and selective suppression of TGF-β isoform expression in cell-based assays and in vivo preclinical models.
Collapse
Affiliation(s)
- Frank Jaschinski
- Isarna Therapeutics. Preclinical Research and Development Department, Leopoldstrasse 254-256, 80807, Munich, Germany
| | | | | |
Collapse
|
64
|
Abstract
Human Epidermal Growth Factor Receptor type 2 (HER2) gene amplification and/or protein overexpression is observed in patients suffering from HER2+ breast cancer. This subtype of breast cancer has improved prognosis due to availability of anti-HER2 therapy. However, drug resistance and tumor recurrence still remains a major concern. Cancer Stem Cells (CSCs) are believed to constitute the subset of cell population that is resistant to drug treatment and possesses characteristics of stem cells. CSCs enable the tumors to thrive despite major insults. This review provides a comprehensive idea about the concept of CSCs in context of HER2+ breast cancer by providing the description of the markers that are used for the identification of CSCs and by elucidating the signaling pathways that are associated with HER2+ breast CSCs. Furthermore, the review also describes the interaction of HER2 with those signaling pathways and the future of targeting CSCs in HER2+ breast cancer.
Collapse
|
65
|
Chen F, Liu X, Bai J, Pei D, Zheng J. The emerging role of RUNX3 in cancer metastasis (Review). Oncol Rep 2015; 35:1227-36. [PMID: 26708741 DOI: 10.3892/or.2015.4515] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/11/2015] [Indexed: 11/06/2022] Open
Abstract
Metastasis remains the major driver of mortality in patients with cancer. The multistep metastatic process starts with the dissemination of tumor cells from a primary site and leading to secondary tumor development in an anatomically distant location. Although significant progress has been made in understanding the molecular characteristics of metastasis, many questions remain regarding the intracellular mechanisms governing transition through the various metastatic stages. The runt-related transcription factor 3 (RUNX3) is a downstream effector of the transforming growth factor-β (TGF-β) signaling pathway, and has critical roles in the regulation of cell death by apoptosis, and in angiogenesis, epithelial-to-mesenchymal transition (EMT), cell migration and invasion. RUNX3 functions as a bona fide initiator of carcinogenesis by linking the Wnt oncogenic and TGF-β tumor suppressive pathways. RUNX3 is frequently inactivated in human cancer cell lines and cancer samples by hemizygous deletion of the Runx3 gene, hypermethylation of the Runx3 promoter, or cytoplasmic sequestration of RUNX3 protein. Inactivation of RUNX3 makes it a putative tumor suppressor in human neoplasia. In the present review, we summarize the proposed roles of RUNX3 in metastasis and, when applicable, highlight the mechanism by which they function.
Collapse
Affiliation(s)
- Feifei Chen
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Dongsheng Pei
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
66
|
Conditioned media from human ovarian cancer endothelial progenitor cells induces ovarian cancer cell migration by activating epithelial-to-mesenchymal transition. Cancer Gene Ther 2015; 22:518-23. [DOI: 10.1038/cgt.2015.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/21/2015] [Accepted: 08/27/2015] [Indexed: 12/27/2022]
|
67
|
Pasquier J, Abu-Kaoud N, Al Thani H, Rafii A. Epithelial to Mesenchymal Transition in a Clinical Perspective. JOURNAL OF ONCOLOGY 2015; 2015:792182. [PMID: 26425122 PMCID: PMC4575734 DOI: 10.1155/2015/792182] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/13/2015] [Indexed: 02/08/2023]
Abstract
Tumor growth and metastatic dissemination rely on cellular plasticity. Among the different phenotypes acquired by cancer cells, epithelial to mesenchymal transition (EMT) has been extensively illustrated. Indeed, this transition allows an epithelial polarized cell to acquire a more mesenchymal phenotype with increased mobility and invasiveness. The role of EMT is quite clear during developmental stage. In the neoplastic context in many tumors EMT has been associated with a more aggressive tumor phenotype including local invasion and distant metastasis. EMT allows the cell to invade surrounding tissues and survive in the general circulation and through a stem cell phenotype grown in the host organ. The molecular pathways underlying EMT have also been clearly defined and their description is beyond the scope of this review. Here we will summarize and analyze the attempts made to block EMT in the therapeutic context. Indeed, till today, most of the studies are made in animal models. Few clinical trials are ongoing with no obvious benefits of EMT inhibitors yet. We point out the limitations of EMT targeting such tumor heterogeneity or the dynamics of EMT during disease progression.
Collapse
Affiliation(s)
- Jennifer Pasquier
- Stem Cell and Microenvironment Laboratory, Department of Genetic Medicine and Obstetrics and Gynecology, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Nadine Abu-Kaoud
- Stem Cell and Microenvironment Laboratory, Department of Genetic Medicine and Obstetrics and Gynecology, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Haya Al Thani
- Stem Cell and Microenvironment Laboratory, Department of Genetic Medicine and Obstetrics and Gynecology, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Arash Rafii
- Stem Cell and Microenvironment Laboratory, Department of Genetic Medicine and Obstetrics and Gynecology, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
68
|
Overcoming EMT-associated resistance to anti-cancer drugs via Src/FAK pathway inhibition. Oncotarget 2015; 5:7328-41. [PMID: 25193862 PMCID: PMC4202126 DOI: 10.18632/oncotarget.2397] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a key process in embryonic development and has been associated with cancer metastasis and drug resistance. For example, in EGFR mutated non-small cell lung cancers (NSCLC), EMT has been associated with acquired resistance to the EGFR inhibitor erlotinib. Moreover, “EGFR-addicted” cancer cell lines induced to undergo EMT become erlotinib-resistant in vitro. To identify potential therapeutic vulnerabilities specifically within these mesenchymal, erlotinib-resistant cells, we performed a small molecule screen of ~200 established anti-cancer agents using the EGFR mutant NSCLC HCC827 cell line and a corresponding mesenchymal derivative line. The mesenchymal cells were more resistant to most tested agents; however, a small number of agents showed selective growth inhibitory activity against the mesenchymal cells, with the most potent being the Abl/Src inhibitor, dasatinib. Analysis of the tyrosine phospho-proteome revealed several Src/FAK pathway kinases that were differentially phosphorylated in the mesenchymal cells, and RNAi depletion of the core Src/FAK pathway components in these mesenchymal cells caused apoptosis. These findings reveal a novel role for Src/FAK pathway kinases in drug resistance and identify dasatinib as a potential therapeutic for treatment of erlotinib resistance associated with EMT.
Collapse
|
69
|
Pan XY, Wang Y, Su J, Huang GX, Cao DM, Qu S, Lu J. The mechanism and significance of synergistic induction of the expression of plasminogen activator inhibitor-1 by glucocorticoid and transforming growth factor beta in human ovarian cancer cells. Mol Cell Endocrinol 2015; 407:37-45. [PMID: 25770462 DOI: 10.1016/j.mce.2015.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 11/17/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) plays a key role in tissue remodeling and tumor development by suppression of plasminogen activator function. Glucocorticoids (GCs) and transforming growth factor beta (TGF-β) signal pathways cross-talk to regulate gene expression, but the mechanism is poorly understood. Here we investigated the mechanism and significance of co-regulation of PAI-1 by TGF-β and dexamethasone (DEX), a synthetic glucocorticoid in ovarian cancer cells. We found that TGF-β and DEX showed rapidly synergistic induction of PAI-1 expression, which contributed to the early pro-adhesion effects. The synergistic induction effect was accomplished by several signal pathways, including GC receptor (GR) pathway and TGF-β-activated p38MAPK, ERK and Smad3 pathways. TGF-β-activated p38MAPK and ERK pathways cross-talked with GR pathway to augment the expression of PAI-1 through enhancing DEX-induced GR phosphorylation at Ser211 in ovarian cancer cells. These findings reveal possible novel mechanisms by which TGF-β pathways cooperatively cross-talk with GR pathway to regulate gene expression.
Collapse
Affiliation(s)
- Xiao-yu Pan
- Department of Endocrinology, Shanghai 10th People's Hospital, Tongji University, Shanghai 200072, China
| | - Yan Wang
- Department of Pathophysiology, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Jie Su
- Department of Pathophysiology, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Gao-xiang Huang
- Department of Pathophysiology, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Dong-mei Cao
- Department of Pathophysiology, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Shen Qu
- Department of Endocrinology, Shanghai 10th People's Hospital, Tongji University, Shanghai 200072, China
| | - Jian Lu
- Department of Pathophysiology, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China.
| |
Collapse
|
70
|
Peng X, Luo Z, Kang Q, Deng D, Wang Q, Peng H, Wang S, Wei Z. FOXQ1 mediates the crosstalk between TGF-β and Wnt signaling pathways in the progression of colorectal cancer. Cancer Biol Ther 2015; 16:1099-109. [PMID: 25955104 DOI: 10.1080/15384047.2015.1047568] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A wide variety of signaling transduction pathways contribute to tumorigenesis. Forkhead box Q1 (FOXQ1) is a member of the forkhead transcription factor family and its upregulation is closely correlated with tumor progression and prognosis of multiple cancer types, including colorectal cancer. However, the molecular mechanisms by which FOXQ1 promotes tumorigenesis, especially cancer cell invasion and metastasis in colorectal cancer, have not been fully elucidated. In the present study, we demonstrate that FOXQ1 is overexpressed in colorectal tumor tissues and its expression level is closely correlated with the stage and lymph node metastasis of colorectal cancer. In in vitro cultured SW480 colorectal cancer cells, knockdown of FOXQ1 expression by small interfering RNA greatly diminished the aggressive tumor behaviors of SW480 cells, including angiogenesis, invasion, epithelial-mesenchymal transition, and resistance to chemotherapy drug-induced apoptosis. Further mechanistic investigation showed that FOXQ1 silencing prevents the nuclear translocation of β-catenin, thus reducing the activity of Wnt signaling. Moreover, TGF-β1 induced the expression of FOXQ1 as well as the migration and invasion of SW480 cells, which was partially prevented following knockdown of FOXQ1. Our results demonstrate that FOXQ1 plays a critical role during the tumorigenesis of colorectal cancer and is a mediator of the crosstalk between Wnt and TGF-β signaling pathways. Our findings provide further insight into the cancer biology of colorectal cancer and suggest that FOXQ1 is a potential therapeutic target for the development of therapies for colorectal cancer.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- 7-AAD, 7-aminoactinomycin D
- DAPI, 4′,6-diamidino-2-phenylindole
- DEPC, Diethy pyrocarbonate
- DMSO, Dimethyl sulfoxide
- EMT, Epithelial-Mesenchymal transition
- FOXQ1
- FOXQ1, Forkhead Box Q1
- L-OHP, Oxaliplatin
- MMP2, Matrix metalloproteinase-2
- MiRNA, MicroRNA
- NC-shRNA, Negative Control-shRNA
- PBS, Phosphate buffer solution
- PBS, phosphate buffered saline
- PKA, proteinkinase A
- PVDF, Polyvinylidene fluoride
- RNAi, RNA interference
- SDS, Sodium dodecyl sulfonate
- TBS, Tris-buffered saline
- TEMED, Tetra methyl ethylene diamine
- TGF-β, Transformin growth β
- TGF-β1
- Tris, Trihydroxymethyl minomethane
- VEGF-A, Vascular endothelial growth factor-A
- Wnt signaling
- aggressive tumor behavior
- cDNA, Complementary DNA
- colorectal cancer
- ddH2O, double distilled H2O
- epithelial-mesenchymal transition
- qRT-PCR, Quantitative real-time PCR
- shRNA, Short hairpin RNA
- μg, Microgramme
- μl, Microliter
Collapse
Affiliation(s)
- Xudong Peng
- a Gastrointestinal Surgical Unit; The First Affiliated Hospital of Chongqing Medical University ; Chongqing , China
| | | | | | | | | | | | | | | |
Collapse
|
71
|
The anticancer effect of (1S,2S,3E,7E,11E)-3,7,11, 15-cembratetraen-17,2-olide(LS-1) through the activation of TGF-β signaling in SNU-C5/5-FU, fluorouracil-resistant human colon cancer cells. Mar Drugs 2015; 13:1340-59. [PMID: 25786063 PMCID: PMC4377987 DOI: 10.3390/md13031340] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 12/28/2022] Open
Abstract
The anticancer effect of (1S,2S,3E,7E,11E)-3,7,11,15-cembratetraen-17,2-olide (LS-1) from Lobophytum sp. has been already reported in HT-29 human colorectal cancer cells. In this study, we examined the effect of LS-1 on the apoptosis induction of SNU-C5/5-FU, fluorouracil-resistant human colon cancer cells. Furthermore, we investigated whether the apoptosis-induction effect of LS-1 could arise from the activation of the TGF-β pathway. In SNU-C5/5-FU treated with LS-1 of 7.1 μM (IC50), we could observe the various apoptotic characteristics, such as the increase of apoptotic bodies, the increase of the sub-G1 hypodiploid cell population, the decrease of the Bcl-2 level, the increase of procaspase-9 cleavage, the increase of procaspase-3 cleavage and the increase of poly(ADP-ribose) polymerase cleavage. Interestingly, the apoptosis-induction effect of LS-1 was also accompanied by the increase of Smad-3 phosphorylation and the downregulation of c-Myc in SNU-C5/5-FU. LS-1 also increased the nuclear localization of phospho-Smad-3 and Smad-4. We examined whether LS-1 could downregulate the expression of carcinoembryonic antigen (CEA), a direct inhibitor of TGF-β signaling. LS-1 decreased the CEA level, as well as the direct interaction between CEA and TGF-βR1 in the apoptosis-induction condition of SNU-C5/5-FU. To examine whether LS-1 can induce apoptosis via the activation of TGF-β signaling, the SNU-C5/5-FU cells were treated with LS-1 in the presence or absence of SB525334, a TGF-βRI kinase inhibitor. SB525334 inhibited the effect of LS-1 on the apoptosis induction. These findings provide evidence demonstrating that the apoptosis-induction effect of LS-1 results from the activation of the TGF-β pathway via the downregulation of CEA in SNU-C5/5-FU.
Collapse
|
72
|
Kim EJ, Kim HJ, Park MK, Kang GJ, Byun HJ, Lee H, Lee CH. Cardamonin Suppresses TGF-β1-Induced Epithelial Mesenchymal Transition via Restoring Protein Phosphatase 2A Expression. Biomol Ther (Seoul) 2015; 23:141-8. [PMID: 25767682 PMCID: PMC4354315 DOI: 10.4062/biomolther.2014.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/12/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022] Open
Abstract
Epithelial mesenchymal transition (EMT) is the first step in metastasis and implicated in the phenotype of cancer stem cells. Therefore, understanding and controlling EMT, are essential to the prevention and cure of metastasis. In the present study, we examined, by Western blot, reverse transcription polymerase chain reaction (RT-PCR), and confocal microscopy, the effects of cardamonin (CDN) on transforming growth factor-β1 (TGF-β1)-induced EMT of A549 lung adenocarcinoma cell lines. TGF-β1 induced expression of N-cadherin and decreased expression of E-cadherin. CDN suppressed N-cadherin expression and restored E-cadherin expression. Further, TGF-β1 induced migration and invasion of A549 cancer cells, which was suppressed by CDN. TGF-β1 induced c-Jun N-terminal kinase (JNK) activation during EMT, but CDN blocked it. Protein serine/threonine phosphatase 2A (PP2A) expression in A549 cancer cells was reduced by TGF-β1 but CDN restored it. The overall data suggested that CDN suppresses TGF-β1-induced EMT via PP2A restoration, making it a potential new drug candidate that controls metastasis.
Collapse
Affiliation(s)
- Eun Ji Kim
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Hyun Ji Kim
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Mi Kyung Park
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Gyeung Jin Kang
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Hyun Jung Byun
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Ho Lee
- National Cancer Center, Goyang 410-769, Republic of Korea
| | - Chang Hoon Lee
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| |
Collapse
|
73
|
Mechanisms of tumor-induced T cell immune suppression and therapeutics to counter those effects. Arch Pharm Res 2015; 38:1415-33. [PMID: 25634101 DOI: 10.1007/s12272-015-0566-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/18/2015] [Indexed: 02/07/2023]
Abstract
The theory of tumor immune surveillance states that the host immune system has means to recognize transformed cells and kills them to prevent growth and spreading of those cells. Nevertheless, cancer cells often survive and outgrow to form a tumor mass and metastasize to other tissues or organs. During the stage of immune evasion of tumor, various changes takes place both in the tumor cells and the tumor microenvironment to divert the anti-tumor immune responses by T cells and natural killer cells. Advances in the basic science in tumor immunology have led to development of many creative strategies to overcome the immune suppression imposed during tumor progression, a few of which have been approved for the treatment of cancer patients in the clinic. In the first part of this review, mechanisms of tumor-induced T cell immune suppression resulting in immune evasion of tumors will be discussed. In the second part, emerging methods to harness the immune responses against tumors will be introduced.
Collapse
|
74
|
Winkler B, Taschik J, Haubitz I, Eyrich M, Schlegel PG, Wiegering V. TGFβ and IL10 have an impact on risk group and prognosis in childhood ALL. Pediatr Blood Cancer 2015; 62:72-9. [PMID: 25263239 DOI: 10.1002/pbc.25142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/12/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cytokines and their genes have been described to have an influence on incidence and prognosis in malignant, infectious and autoimmune disease. We previously described the impact of cytokine production on prognosis in paediatric standard-risk acute lymphoblastic leukaemia (ALL). PROCEDURE In this study, we investigated the influence of cytokine gene polymorphisms (TNFα, TGFβ, IL10 and IFNγ) on frequency, risk group and prognosis in 95 paediatric ALL-patients. We further report on intracellular production of these cytokines in T-cells. RESULTS IL10 high-producer-haplotypes were reduced in ALL-patients compared with healthy controls and resulted in a reduced relapse rate compared with low-producer haplotypes. TGFβ high-producer-haplotypes were correlated with a high initial blast-count (codon 25: G/G) and were elevated in high-risk ALL-patients (codon 10: T/T). IL10 was positively and IFNγ-production was negatively correlated with initial blast-count. At diagnosis the expression of TNFα and IFNγ was reduced in patients compared with healthy controls. This was more pronounced in high-risk and in T-ALL-patients. CONCLUSION We conclude that gene-polymorphisms of the regulatory/anti-inflammatory cytokines, TGFβ and IL10, but not of the pro-inflammatory cytokines, IFNγ and TNFα, have an impact on prognosis and risk-group of ALL. However, the reduced capacity to produce pro-inflammatory cytokines at diagnosis may serve as another important, functional risk factor. These data may help in further risk stratification and adaptation of therapy-intensity in paediatric patients with ALL.
Collapse
Affiliation(s)
- B Winkler
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Würzburg, Children's Hospital, Germany
| | | | | | | | | | | |
Collapse
|
75
|
Loomans HA, Andl CD. Intertwining of Activin A and TGFβ Signaling: Dual Roles in Cancer Progression and Cancer Cell Invasion. Cancers (Basel) 2014; 7:70-91. [PMID: 25560921 PMCID: PMC4381251 DOI: 10.3390/cancers7010070] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/23/2014] [Indexed: 12/22/2022] Open
Abstract
In recent years, a significant amount of research has examined the controversial role of activin A in cancer. Activin A, a member of the transforming growth factor β (TGFβ) superfamily, is best characterized for its function during embryogenesis in mesoderm cell fate differentiation and reproduction. During embryogenesis, TGFβ superfamily ligands, TGFβ, bone morphogenic proteins (BMPs) and activins, act as potent morphogens. Similar to TGFβs and BMPs, activin A is a protein that is highly systemically expressed during early embryogenesis; however, post-natal expression is overall reduced and remains under strict spatiotemporal regulation. Of importance, normal post-natal expression of activin A has been implicated in the migration and invasive properties of various immune cell types, as well as endometrial cells. Aberrant activin A signaling during development results in significant morphological defects and premature mortality. Interestingly, activin A has been found to have both oncogenic and tumor suppressor roles in cancer. Investigations into the role of activin A in prostate and breast cancer has demonstrated tumor suppressive effects, while in lung and head and neck squamous cell carcinoma, it has been consistently shown that activin A expression is correlated with increased proliferation, invasion and poor patient prognosis. Activin A signaling is highly context-dependent, which is demonstrated in studies of epithelial cell tumors and the microenvironment. This review discusses normal activin A signaling in comparison to TGFβ and highlights how its dysregulation contributes to cancer progression and cell invasion.
Collapse
Affiliation(s)
- Holli A Loomans
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Claudia D Andl
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
76
|
Abstract
Tubulointerstitial fibrosis and glomerulosclerosis, are a major feature of end stage chronic kidney disease (CKD), characterised by an excessive accumulation of extracellular matrix (ECM) proteins. Transforming growth factor beta-1 (TGF-β1) is a cytokine with an important role in many steps of renal fibrosis such as myofibroblast activation and proliferation, ECM protein synthesis and inflammatory cell infiltration. Endoglin is a TGF-β co-receptor that modulates TGF-β responses in different cell types. In numerous cells types, such as mesangial cells or myoblasts, endoglin regulates negatively TGF-β-induced ECM protein expression. However, recently it has been demonstrated that 'in vivo' endoglin promotes fibrotic responses. Furthermore, several studies have demonstrated an increase of endoglin expression in experimental models of renal fibrosis in the kidney and other tissues. Nevertheless, the role of endoglin in renal fibrosis development is unclear and a question arises: Does endoglin protect against renal fibrosis or promotes its development? The purpose of this review is to critically analyse the recent knowledge relating to endoglin and renal fibrosis. Knowledge of endoglin role in this pathology is necessary to consider endoglin as a possible therapeutic target against renal fibrosis.
Collapse
|
77
|
Koh M, Woo Y, Valiathan RR, Jung HY, Park SY, Kim YN, Kim HRC, Fridman R, Moon A. Discoidin domain receptor 1 is a novel transcriptional target of ZEB1 in breast epithelial cells undergoing H-Ras-induced epithelial to mesenchymal transition. Int J Cancer 2014; 136:E508-20. [PMID: 25155634 DOI: 10.1002/ijc.29154] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 07/31/2014] [Accepted: 08/14/2014] [Indexed: 01/12/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT) process allows carcinoma cells to dissociate from the primary tumor thereby facilitating tumor cell invasion and metastasis. Ras-dependent hyperactive signaling is commonly associated with tumorigenesis, invasion, EMT, and metastasis. However, the downstream effectors by which Ras regulates EMT remain ill defined. In this study, we show that the H-Ras pathway leads to mesenchymal-like phenotypic changes in human breast epithelial cells by controlling the ZEB1/microRNA-200c axis. Moreover, H-Ras suppresses the expression of the discoidin domain receptor 1 (DDR1), a collagen receptor tyrosine kinase, via ZEB1, thus identifying ZEB1 as a novel transcriptional repressor of DDR1. Mutation studies on the putative promoter of the DDR1 gene revealed that bipartite Z- and E-box elements play a key role in transcriptional repression of DDR1 in Hs578T and MDA-MB-231 breast carcinoma cell lines by ZEB1. Furthermore, we found an inverse correlation between ZEB1 and DDR1 expression in various cancer cell lines and in human breast carcinoma tissues. Consistently, overexpression of DDR1 reduced the invasive phenotype of mesenchymal-like triple-negative breast cancer cells in 3D cultures and in vivo. Thus, ZEB1's role in maintenance of EMT in breast carcinoma cells is mediated in part by its ability to suppress DDR1 expression and consequently contribute to the activation of the invasive phenotype. Taken together, our results unveil a novel H-Ras/ZEB1/DDR1 network that contributes to breast cancer progression in triple-negative breast cancers.
Collapse
Affiliation(s)
- Minsoo Koh
- College of Pharmacy, Duksung Women's University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Zarzynska JM. Two faces of TGF-beta1 in breast cancer. Mediators Inflamm 2014; 2014:141747. [PMID: 24891760 PMCID: PMC4033515 DOI: 10.1155/2014/141747] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/16/2014] [Accepted: 04/24/2014] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) is potentially life-threatening malignancy that still causes high mortality among women. Scientific research in this field is focused on deeper understanding of pathogenesis and progressing of BC, in order to develop relevant diagnosis and improve therapeutic treatment. Multifunctional cytokine TGF- β 1 is one of many factors that have a direct influence on BC pathophysiology. Expression of TGF- β 1, induction of canonical and noncanonical signaling pathways, and mutations in genes encoding TGF- β 1 and its receptors are correlated with oncogenic activity of this cytokine. In early stages of BC this cytokine inhibits epithelial cell cycle progression and promotes apoptosis, showing tumor suppressive effects. However, in late stages, TGF- β 1 is linked with increased tumor progression, higher cell motility, cancer invasiveness, and metastasis. It is also involved in cancer microenvironment modification and promotion of epithelial to mesenchymal transition (EMT). This review summarizes the current knowledge on the phenomenon called "TGF- β 1 paradox", showing that better understanding of TGF- β 1 functions can be a step towards development of new therapeutic approaches. According to current knowledge several drugs against TGF- β 1 have been developed and are either in nonclinical or in early stages of clinical investigation.
Collapse
Affiliation(s)
- Joanna Magdalena Zarzynska
- Department of Food Hygiene and Public Health, Faculty of Veterinary Medicine, WULS-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| |
Collapse
|
79
|
Gudey SK, Wallenius A, Landström M. Regulated intramembrane proteolysis of the TGFβ type I receptor conveys oncogenic signals. Future Oncol 2014; 10:1853-61. [PMID: 24597658 DOI: 10.2217/fon.14.45] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cancer cells produce high levels of TGFβ, a multipotent cytokine. Binding of TGFβ to its cell surface receptors, the transmembrane serine/threonine kinases TβRII and TβRI, causes phosphorylation and activation of intracellular latent Smad transcription factors. Nuclear Smads act in concert with specific transcription factors to reprogram epithelial cells to become invasive mesenchymal cells. TGFβ also propagates non-canonical signals, so it is crucial to have a better understanding of the underlying molecular mechanisms which favor this pathway. Here we highlight our recent discovery that TGFβ promotes the proteolytic cleavage of TβRI in cancer cells, resulting in the liberation and nuclear translocation of its intracellular domain, acting as co-regulator to transcribe pro-invasive genes. This newly identified oncogenic TGFβ pathway resembles the Notch signaling pathway. We discuss our findings in relation to Notch and provide a short overview of other growth factors that transduce signals via nuclear translocation of their cell surface receptors.
Collapse
Affiliation(s)
- Shyam Kumar Gudey
- Department of Medical Biosciences, Pathology, Umeå University, SE-901 85 Umeå, Sweden
| | | | | |
Collapse
|