1
|
Zhao Y, Jin LJ, Zhang XY. Exosomal miRNA-205 promotes breast cancer chemoresistance and tumorigenesis through E2F1. Aging (Albany NY) 2021; 13:18498-18514. [PMID: 34292880 PMCID: PMC8351670 DOI: 10.18632/aging.203298] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/19/2021] [Indexed: 01/09/2023]
Abstract
Breast cancer (BC) is a common malignant tumor in females. The challenge in treating BC is overcoming chemoresistance. Exosome-mediated transfer of miRNAs is a molecule-shuttle in intercellular communication. Thus, we aimed to investigate whether exosomal miRNA-205 could affect chemoresistance and tumorigenesis in recipient tumor cells and to elucidate the underlying mechanism in vivo and in vitro. Microarray and qRT-PCR assays demonstrated that miRNA-205 was upregulated in tamoxifen resistance MCF-7/TAMR-1 (M/T) cells and M/T cell-derived exosomes (M/T-Exo). The M/T-Exo was internalized by human BC cells (BCCs), causing increased expression of miRNA-205 in BCCs. Coculturing with M/T-Exo promoted tamoxifen resistance, proliferation, migration, and invasion while suppressed apoptosis in recipient BCCs, which were associated with activating the caspase pathway and phosphorylating Akt. Luciferase reporter assays showed that miRNA-205 directly targeted E2F Transcription Factor 1 (E2F1) in BCCs. Furthermore, knockdown of miRNA-205 or overexpression of E2F1 reversed the roles of M/T-Exo in BCCs. In vivo experiments showed that the intratumoral injection of M/T-Exo caused greater tamoxifen resistance and larger tumor size relative to mice treated with miRNA-205-knockdown or E2F1-overexpressing BCCs. Together, the results suggest that exosomal miRNA-205 may promote tamoxifen resistance and tumorigenesis in BC through targeting E2F1 in vivo and in vitro.
Collapse
Affiliation(s)
- Yan Zhao
- Thyroid and Breast Department, Extra-Thyroid and Breast Neoplasms, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Li-Jun Jin
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Xiao-Yu Zhang
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
2
|
Chang YC, Oram MK, Bielinsky AK. SUMO-Targeted Ubiquitin Ligases and Their Functions in Maintaining Genome Stability. Int J Mol Sci 2021; 22:ijms22105391. [PMID: 34065507 PMCID: PMC8161396 DOI: 10.3390/ijms22105391] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Small ubiquitin-like modifier (SUMO)-targeted E3 ubiquitin ligases (STUbLs) are specialized enzymes that recognize SUMOylated proteins and attach ubiquitin to them. They therefore connect the cellular SUMOylation and ubiquitination circuits. STUbLs participate in diverse molecular processes that span cell cycle regulated events, including DNA repair, replication, mitosis, and transcription. They operate during unperturbed conditions and in response to challenges, such as genotoxic stress. These E3 ubiquitin ligases modify their target substrates by catalyzing ubiquitin chains that form different linkages, resulting in proteolytic or non-proteolytic outcomes. Often, STUbLs function in compartmentalized environments, such as the nuclear envelope or kinetochore, and actively aid in nuclear relocalization of damaged DNA and stalled replication forks to promote DNA repair or fork restart. Furthermore, STUbLs reside in the same vicinity as SUMO proteases and deubiquitinases (DUBs), providing spatiotemporal control of their targets. In this review, we focus on the molecular mechanisms by which STUbLs help to maintain genome stability across different species.
Collapse
|
3
|
Mintis DG, Chasapi A, Poulas K, Lagoumintzis G, Chasapis CT. Assessing the Direct Binding of Ark-Like E3 RING Ligases to Ubiquitin and Its Implication on Their Protein Interaction Network. Molecules 2020; 25:molecules25204787. [PMID: 33086510 PMCID: PMC7594095 DOI: 10.3390/molecules25204787] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022] Open
Abstract
The ubiquitin pathway required for most proteins’ targeted degradation involves three classes of enzymes: E1-activating enzyme, E2-conjugating enzyme, and E3-ligases. The human Ark2C is the single known E3 ligase that adopts an alternative, Ub-dependent mechanism for the activation of Ub transfer in the pathway. Its RING domain binds both E2-Ub and free Ub with high affinity, resulting in a catalytic active UbR-RING-E2-UbD complex formation. We examined potential changes in the conformational plasticity of the Ark2C RING domain and its ligands in their complexed form within the ubiquitin pathway through molecular dynamics (MD). Three molecular mechanics force fields compared to previous NMR relaxation studies of RING domain of Arkadia were used for effective and accurate assessment of MDs. Our results suggest the Ark2C Ub-RING docking site has a substantial impact on maintaining the conformational rigidity of E2-E3 assembly, necessary for the E3’s catalytic activity. In the UbR-RING-E2-UbD catalytic complex, the UbR molecule was found to have greater mobility than the other Ub, bound to E2. Furthermore, network-based bioinformatics helped us identify E3 RING ligase candidates which potentially exhibit similar structural modules as Ark2C, along with predicted substrates targeted by the Ub-binding RING Ark2C. Our findings could trigger a further exploration of related unrevealed functions of various other E3 RING ligases.
Collapse
Affiliation(s)
- Dimitris G. Mintis
- Laboratory of Statistical Thermodynamics and Macromolecules, Department of Chemical Engineering, University of Patras & FORTH/ICE-HT, 26504 Patras, Greece;
| | - Anastasia Chasapi
- Biological Computation & Process Lab, Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, 57001 Thessaloniki, Greece;
| | - Konstantinos Poulas
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26504 Patras, Greece;
- Institute of Research and Innovation-IRIS, Patras Science Park SA, Stadiou, Platani, Rio, 26504 Patras, Greece
| | - George Lagoumintzis
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26504 Patras, Greece;
- Institute of Research and Innovation-IRIS, Patras Science Park SA, Stadiou, Platani, Rio, 26504 Patras, Greece
- Correspondence: (G.L.); (C.T.C.); Tel.: +30-2610-996-312 (G.L.); +30-2610-996-261 (C.T.C.)
| | - Christos T. Chasapis
- NMR Center, Instrumental Analysis Laboratory, School of Natural Sciences, University of Patras, 26504 Patras, Greece
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology, Hellas (FORTH/ICE-HT), 26504 Patras, Greece
- Correspondence: (G.L.); (C.T.C.); Tel.: +30-2610-996-312 (G.L.); +30-2610-996-261 (C.T.C.)
| |
Collapse
|
4
|
Li P, Wang QS, Zhai Y, Xiong RP, Chen X, Liu P, Peng Y, Zhao Y, Ning YL, Yang N, Zhou YG. Ski mediates TGF-β1-induced fibrosarcoma cell proliferation and promotes tumor growth. J Cancer 2020; 11:5929-5940. [PMID: 32922535 PMCID: PMC7477421 DOI: 10.7150/jca.46074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/20/2020] [Indexed: 11/05/2022] Open
Abstract
Background: TGF-β1 promotes cell proliferation in only some tumors and exerts bidirectional regulatory effects on the proliferation of fibroblasts. This study intends to explore whether the mechanism is related to increased expression of Ski. Methods: Cell proliferation of the fibrosarcoma cell line L929 was assessed with an ELISA BrdU kit. The mRNA and protein expression levels of the corresponding factors were measured by RT-qPCR, immunohistochemistry or Western blotting in vitro and in vivo. Additionally, c-Ski was knocked down using RNAi. The expression of Ski in human dermatofibrosarcoma protuberans (DFSP) specimens was measured by immunohistochemistry. Results: TGF-β1 promoted the continued proliferation of L929 cells in a dose-dependent manner, with increased c-Ski expression levels. Conversely, inhibition of c-Ski significantly abrogated this unidirectional effect, significantly inhibited the decrease in p21 protein levels and did not affect the increase in p-Smad2/3 levels upon TGF-β1 treatment. Similarly, inhibition of c-Ski significantly abrogated the growth-promoting effect of TGF-β1 on xenograft tumors. Furthermore, we found that high expression of Ski in DFSP was correlated with a low degree of tumor differentiation. Conclusions: Our data reveal that high c-Ski expression is a cause of TGF-β1-promoted proliferation in fibrosarcoma tumor cells and show that inhibiting Ski expression might be effective for treating tumors with high Ski levels.
Collapse
Affiliation(s)
- Ping Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Qiu-Shi Wang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China.,Department of Pathology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Yu Zhai
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Ren-Ping Xiong
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Xing Chen
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Ping Liu
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Yan Peng
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Ya-Lei Ning
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Nan Yang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Yuan-Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, People's Republic of China
| |
Collapse
|
5
|
Zeglinski MR, Moghadam AR, Ande SR, Sheikholeslami K, Mokarram P, Sepehri Z, Rokni H, Mohtaram NK, Poorebrahim M, Masoom A, Toback M, Sareen N, Saravanan S, Jassal DS, Hashemi M, Marzban H, Schaafsma D, Singal P, Wigle JT, Czubryt MP, Akbari M, Dixon IM, Ghavami S, Gordon JW, Dhingra S. Myocardial Cell Signaling During the Transition to Heart Failure. Compr Physiol 2018; 9:75-125. [DOI: 10.1002/cphy.c170053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
6
|
Hou F, Liu RX, Yin CH. Arkadia: Characteristics, function and role in development of human diseases. Shijie Huaren Xiaohua Zazhi 2016; 24:3963-3969. [DOI: 10.11569/wcjd.v24.i28.3963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ubiquitination of proteins is a post-translational modification that involves targeting and degrading misfolded or unwanted proteins by the proteasome. Arkadia, a RING-type E3 ubiquitin ligase also known as RNF111, confers the substrate specificity for ubiquitination and has a pivotal role in catalyzing the degradation of key signaling molecules. Recent research reveals that Arkadia plays a pivotal role in the transforming growth factor-β1 signaling pathway by catalyzing the degradation of key signaling molecules. In this review, we highlight the recent progress in understanding the characteristics, function and the role of Arkadia in the development of human diseases.
Collapse
|
7
|
Kim H, Cho YM, Ko YG, Choe C, Seong HH. Relationship between Sloan-Kettering virus expression and mammalian follicular development. ZYGOTE 2015:1-9. [PMID: 26228242 DOI: 10.1017/s0967199415000362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Sloan-Kettering virus gene, a product of a cellular proto-oncogene c-Ski is a unique nuclear pro-oncoprotein and belongs to the Ski/Sno proto-oncogene family. The aim of the present study was to locate Ski protein in rat ovaries in order to find insights into the possible involvement of Ski in follicular development. First, expression of c-Ski mRNA in the ovaries of adult female rats was confirmed by RT-PCR. Then, ovaries obtained on the day of estrus were subjected to immunohistochemical analysis for Ski and proliferating cell nuclear antigen (PCNA) in combination with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). RT-PCR and in situ hybridization revealed that c-Ski mRNA was expressed in the ovaries of the adult rat on the day of estrous and localized mainly in the granulose cells. Ski was expressed in granulosa cells that were positive for TUNEL, but negative for PCNA, regardless of the shape and size of follicles. Expression of Ski in TUNEL-positive granulosa cells, but not in PCNA-positive granulosa cells, was also verified in rats having atretic follicles with double staining. These results indicate that Ski is profoundly expressed in the granulosa cells of atretic follicles, but not in growing follicles. Based on the present findings, Ski may play a role in the apoptosis of granulosa cells during follicular atresia.
Collapse
Affiliation(s)
- Hyun Kim
- Department of Veterinary Physiology,Graduate School of Agricultural and Life Science,The University of Tokyo,1-1-1 Yayoi,Bunkyo-ku,Tokyo 113-8657,Japan
| | - Young Moo Cho
- Animal Genetic Resources Research Center,National Institute of Animal Science,RDA,Namwon 590-832,Republic of Korea
| | - Yeoung-Gyu Ko
- Animal Biotechnology Division,National Institute of Animal Science,RDA,Suwon 441-706,Republic of Korea
| | - Changyong Choe
- Animal Genetic Resources Research Center,National Institute of Animal Science,RDA,Namwon 590-832,Republic of Korea
| | - Hwan-Hoo Seong
- Animal Genetic Resources Research Center,National Institute of Animal Science,RDA,Namwon 590-832,Republic of Korea
| |
Collapse
|
8
|
Chen H, Yang T, Lei Z, Wang L, Yang H, Tong X, Yang WT, Zhao J, Gu Y, Chen Y, Zhang HT. RNF111/Arkadia is regulated by DNA methylation and affects TGF-β/Smad signaling associated invasion in NSCLC cells. Lung Cancer 2015; 90:32-40. [PMID: 26238425 DOI: 10.1016/j.lungcan.2015.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 12/30/2022]
Abstract
OBJECTIVES RNF111/Arkadia is a critical regulator of TGF-β signaling, being required for SMAD3-mediated responses such as TGF-β-induced repression of E-cadherin. Previous studies show that mutations in RNF111 in human cancers are rare and RNF111 promotes lung tumor metastasis. However, the epigenetic mechanisms underlying the role of RNF111 in non-small cell lung cancer (NSCLC) metastasis remain unknown. Here, we mainly focused on low- (95C) and high-metastatic (95D) NSCLC cell lines, which share a similar genetic background, and investigated the methylation-based regulation of RNF111 expression. MATERIALS AND METHODS Clonal bisulfite sequencing, real-time qRT-PCR, western blot analysis, luciferase reporter assays, RNA interference, chromatin immunoprecipitation (ChIP) assay and transwell migration and invasion assays were performed on human NSCLC cell lines 95C and 95D. RESULTS RNF111 was high-expressed in 95D cells, which showed low-level methylation at -459CpG site in RNF111 promoter. The opposite results were obtained in 95C cells. Cell-based and biochemical assays revealed that -459CpG methylation can inhibit RNF111 transcriptional expression by interfering with the recruitment of Sp1 to RNF111 promoter. On TGF-β stimulation, siRNA-mediated RNF111 knockdown inhibited TGF-β/Smad signaling activity and Snail (an inducer of metastasis) expression, and enhanced E-cadherin (an epithelial-to-mesenchymal transition marker) expression in 95C and 95D cells. Furthermore, demethylation-induced upregulation of RNF111 enhanced phosphorylation of SMAD3 and Snail expression, and repressed E-cadherin expression in 95C cells expressing low RNF111. CONCLUSIONS Our results suggest that -459CpG methylation in Sp1-binding site of RNF111 promoter transcriptionally decreases RNF111 expression, which inhibits TGF-β/Smad signaling associated invasion in NSCLC cells.
Collapse
Affiliation(s)
- Hongbing Chen
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China
| | - Tianjie Yang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China
| | - Zhe Lei
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China
| | - Longqiang Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China
| | - Haiping Yang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China
| | - Xin Tong
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China
| | - Wen-Tao Yang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou 215004, China
| | - Jun Zhao
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou 215006, China
| | - Yunbin Gu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou 215006, China
| | - Yongbing Chen
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou 215004, China.
| | - Hong-Tao Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
9
|
Motizuki M, Saitoh M, Miyazawa K. Maid is a negative regulator of transforming growth factor-β-induced cell migration. J Biochem 2015; 158:435-44. [PMID: 26002959 DOI: 10.1093/jb/mvv054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/16/2015] [Indexed: 12/19/2022] Open
Abstract
Maternal Id-like molecule (Maid) is a dominant negative helix-loop-helix protein that has been implicated in regulating gene expression as well as cell-cycle progression. Overexpressed Maid was previously shown to inhibit certain cellular responses induced by transforming growth factor-β (TGF-β), such as TGF-β-induced cytostasis and cell motility, but not epithelial-mesenchymal transition (EMT). The role of endogenous Maid in regulating TGF-β signalling, however, has not been elucidated. We have found evidence that endogenous Maid negatively regulates TGF-β-induced cell motility. Maid knockdown enhanced TGF-β-induced cell motility as measured by chamber migration and wound healing assays but did not affect cell motility induced by bone morphogenetic protein (BMP)-4. Endogenous Maid does not appear to be involved in regulating TGF-β-induced cytostasis, resistance to apoptosis or EMT. Notably, Maid expression was induced in the delayed phase (later than 24 h) after TGF-β stimulation whereas the expression of two other negative feedback regulators, Smad7 and SnoN, was induced as early as 1 h after stimulation. These findings indicate that Maid is a unique negative feedback regulator of TGF-β signalling in its mode of action as well as the timing of its induction.
Collapse
Affiliation(s)
- Mitsuyoshi Motizuki
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Masao Saitoh
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Keiji Miyazawa
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
10
|
Tsubakihara Y, Hikita A, Yamamoto S, Matsushita S, Matsushita N, Oshima Y, Miyazawa K, Imamura T. Arkadia enhances BMP signalling through ubiquitylation and degradation of Smad6. J Biochem 2015; 158:61-71. [PMID: 25762727 DOI: 10.1093/jb/mvv024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/17/2015] [Indexed: 02/02/2023] Open
Abstract
Arkadia, a positive regulator of Smad-dependent signalling via the transforming growth factor-β (TGF-β) family, is an E3 ubiquitin ligase that induces ubiquitylation and proteasome-dependent degradation of TGF-β suppressors such as Smad7, c-Ski and SnoN. In this study, we examined the effects of Arkadia on bone morphogenetic protein (BMP)-induced osteoblast differentiation. Knockdown of Arkadia reduced mineralization and expression of osteoblast differentiation markers. Furthermore, we showed that Smad6, a BMP-specific inhibitory Smad, is a target of Arkadia: wild-type (WT) Arkadia, but not the C937A (CA) mutant lacking E3 ubiquitin-ligase activity, induced ubiquitylation and proteasome-dependent degradation of Smad6. Accordingly, protein levels of Smad6, Smad7 and c-Ski were elevated in MEFs from Arkadia KO mice. Finally, expression of Arkadia attenuated blockade of BMP signalling by Smad6 in a transcriptional reporter assay. These results demonstrate that Smad6 is a novel target of Arkadia, and that Arkadia positively regulates BMP signalling via degradation of Smad6, Smad7 and c-Ski/SnoN.
Collapse
Affiliation(s)
- Yutaro Tsubakihara
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Atsuhiko Hikita
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shin Yamamoto
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Sachi Matsushita
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Natsuki Matsushita
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yusuke Oshima
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-im
| | - Keiji Miyazawa
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-im
| |
Collapse
|
11
|
Imamura T, Oshima Y, Hikita A. Regulation of TGF-β family signalling by ubiquitination and deubiquitination. J Biochem 2013; 154:481-9. [PMID: 24165200 DOI: 10.1093/jb/mvt097] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Members of the transforming growth factor-β (TGF-β) family, including TGF-βs, activin and bone morphogenetic proteins (BMPs), are multifunctional proteins that regulate a wide variety of cellular responses, such as proliferation, differentiation, migration and apoptosis. TGF-β family signalling is mainly mediated by membranous serine/threonine kinase receptors and intracellular Smad proteins. This signalling is tightly regulated by various post-translational modifications including ubiquitination. Several E3 ubiquitin ligases play a crucial role in the recognition and ubiquitin-dependent degradation of TGF-β family receptors, Smad proteins and their interacted proteins to regulate positively and negatively TGF-β family signalling. In contrast, non-degradative ubiquitin modifications also regulate TGF-β family signalling. Recently, in addition to protein ubiquitination, deubiquitination by deubiquitinating enzymes has been reported to control TGF-β family signalling pathways. Interestingly, more recent studies suggest that TGF-β signalling is not only regulated via ubiquitination and/or deubiquitination, but also it relies on ubiquitination for its effect on other pathways. Thus, ubiquitin modifications play key roles in TGF-β family signal transduction and cross-talk between TGF-β family signalling and other signalling pathways. Here, we review the current understandings of the positive and negative regulatory mechanisms by ubiquitin modifications that control TGF-β family signalling.
Collapse
Affiliation(s)
- Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine; Division of Bio-imaging, Proteo-Science Center, Ehime University; Translational Research Center, Ehime University Hospital; and Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Shitsukawa, Toon, Ehime 791-0295, Japan
| | | | | |
Collapse
|
12
|
Al-Salihi MA, Herhaus L, Sapkota GP. Regulation of the transforming growth factor β pathway by reversible ubiquitylation. Open Biol 2013; 2:120082. [PMID: 22724073 PMCID: PMC3376735 DOI: 10.1098/rsob.120082] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 04/25/2012] [Indexed: 12/20/2022] Open
Abstract
The transforming growth factor β (TGFβ) signalling pathway plays a central role during embryonic development and in adult tissue homeostasis. It regulates gene transcription through a signalling cascade from cell surface receptors to intracellular SMAD transcription factors and their nuclear cofactors. The extent, duration and potency of signalling in response to TGFβ cytokines are intricately regulated by complex biochemical processes. The corruption of these regulatory processes results in aberrant TGFβ signalling and leads to numerous human diseases, including cancer. Reversible ubiquitylation of pathway components is a key regulatory process that plays a critical role in ensuring a balanced response to TGFβ signals. Many studies have investigated the mechanisms by which various E3 ubiquitin ligases regulate the turnover and activity of TGFβ pathway components by ubiquitylation. Moreover, recent studies have shed new light into their regulation by deubiquitylating enzymes. In this report, we provide an overview of current understanding of the regulation of TGFβ signalling by E3 ubiquitin ligases and deubiquitylases.
Collapse
Affiliation(s)
- Mazin A Al-Salihi
- Medical Research Council-Protein Phosphorylation Unit, Sir James Black Centre, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | | | | |
Collapse
|
13
|
Ski protein levels increase during in vitro progression of HPV16-immortalized human keratinocytes and in cervical cancer. Virology 2013; 444:100-8. [PMID: 23809940 DOI: 10.1016/j.virol.2013.05.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/23/2013] [Accepted: 05/30/2013] [Indexed: 01/23/2023]
Abstract
We compared the levels of the Ski oncoprotein, an inhibitor of transforming growth factor-beta (TGF-β) signaling, in normal human keratinocytes (HKc), HPV16 immortalized HKc (HKc/HPV16), and differentiation resistant HKc/HPV16 (HKc/DR) in the absence and presence of TGF-β. Steady-state Ski protein levels increased in HKc/HPV16 and even further in HKc/DR, compared to HKc. TGF-β treatment of HKc, HKc/HPV16, and HKc/DR dramatically decreased Ski. TGF-β-induced Ski degradation was delayed in HKc/DR. Ski and phospho-Ski protein levels are cell cycle dependent with maximal Ski expression and localization to centrosomes and mitotic spindles during G2/M. ShRNA knock down of Ski in HKc/DR inhibited cell proliferation. More intense nuclear and cytoplasmic Ski staining and altered Ski localization were found in cervical cancer samples compared to adjacent normal tissue in a cervical cancer tissue array. Overall, these studies demonstrate altered Ski protein levels, degradation and localization in HPV16-transformed human keratinocytes and in cervical cancer.
Collapse
|
14
|
Motizuki M, Isogaya K, Miyake K, Ikushima H, Kubota T, Miyazono K, Saitoh M, Miyazawa K. Oligodendrocyte transcription factor 1 (Olig1) is a Smad cofactor involved in cell motility induced by transforming growth factor-β. J Biol Chem 2013; 288:18911-22. [PMID: 23720758 DOI: 10.1074/jbc.m113.480996] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Transforming growth factor (TGF)-β plays crucial roles in embryonic development and adult tissue homeostasis by eliciting various cellular responses in target cells. TGF-β signaling is principally mediated through receptor-activated Smad proteins, which regulate expression of target genes in cooperation with other DNA-binding transcription factors (Smad cofactors). In this study, we found that the basic helix-loop-helix transcription factor Olig1 is a Smad cofactor involved in TGF-β-induced cell motility. Knockdown of Olig1 attenuated TGF-β-induced cell motility in chamber migration and wound healing assays. In contrast, Olig1 knockdown had no effect on bone morphogenetic protein-induced cell motility, TGF-β-induced cytostasis, or epithelial-mesenchymal transition. Furthermore, we observed that cooperation of Smad2/3 with Olig1 is regulated by a peptidyl-prolyl cis/trans-isomerase, Pin1. TGF-β-induced cell motility, induction of Olig1-regulated genes, and physical interaction between Smad2/3 and Olig1 were all inhibited after knockdown of Pin1, indicating a novel mode of regulation of Smad signaling. We also found that Olig1 interacts with the L3 loop of Smad3. Using a synthetic peptide corresponding to the L3 loop of Smad3, we succeeded in selectively inhibiting TGF-β-induced cell motility. These findings may lead to a new strategy for selective regulation of TGF-β-induced cellular responses.
Collapse
Affiliation(s)
- Mitsuyoshi Motizuki
- Department of Biochemistry, University of Yamanashi, Yamanashi 409-3898, Japan
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Katsuno Y, Ehata S, Yashiro M, Yanagihara K, Hirakawa K, Miyazono K. Coordinated expression of REG4 and aldehyde dehydrogenase 1 regulating tumourigenic capacity of diffuse-type gastric carcinoma-initiating cells is inhibited by TGF-β. J Pathol 2012; 228:391-404. [PMID: 22430847 DOI: 10.1002/path.4020] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/05/2012] [Accepted: 03/09/2012] [Indexed: 12/14/2022]
Abstract
Aldehyde dehydrogenase 1 (ALDH1) has been shown to serve as a marker for cancer-initiating cells (CICs), but little is known about the regulation of the CIC functions of ALDH1+ cancer cells. We isolated ALDH1+ cells from human diffuse-type gastric carcinoma cells and characterized these cells using an Aldefluor assay. ALDH1+ cells constituted 5-8% of the human diffuse-type gastric carcinoma cells, OCUM-2MLN and HSC-39; were more tumourigenic than ALDH1- cells; and were able to self-renew and generate heterogeneous cell populations. Using gene expression microarray analyses, we identified REG4 (regenerating islet-derived family, member 4) as one of the genes up-regulated in ALDH1+ cells, and thus as a novel marker for ALDH1+ tumour cells. Induced expression of REG4 enhanced the colony-forming ability of OCUM-2MLN cells, while knockdown of REG4 inhibited the tumourigenic potential of ALDH1+ cells. We further found that TGF-β signalling reduces the expression of ALDH1 and REG4, and the size of the ALDH1+ cell population. In human diffuse-type gastric carcinoma tissues, the expression of ALDH1 and REG4 correlated with each other, as assessed by immunohistochemistry, and ALDH1 expression correlated inversely with Smad3 phosphorylation as a measure of TGF-β signalling. These findings illustrate that, in diffuse-type gastric carcinoma, REG4 is up-regulated in ALDH1+ CICs, and that the increased tumourigenic ability of ALDH1+ cells depends on REG4. Moreover, TGF-β down-regulates ALDH1 and REG4 expression, which correlates with a reduction in CIC population size and tumourigenicity. Targeting REG4 in ALDH1+ CICs may provide a novel strategy in the treatment of diffuse-type gastric carcinoma.
Collapse
Affiliation(s)
- Yoko Katsuno
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Kim H, Kim DH, Park SB, Ko YG, Kim SW, Do YJ, Park JH, Yang BS. Induction of Ski Protein Expression upon Luteinization in Rat Granulosa Cells. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2012; 25:635-41. [PMID: 25049607 PMCID: PMC4093106 DOI: 10.5713/ajas.2011.11336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 02/02/2012] [Accepted: 11/23/2011] [Indexed: 12/14/2022]
Abstract
Ski protein is implicated in proliferation/differentiation in a variety of cells. We had previously reported that Ski protein is present in granulosa cells of atretic follicles, but not in preovulatory follicles, suggesting that Ski has a role in apoptosis of granulosa cells. The alternative fate of granulosa cells other than apoptosis is to differentiate to luteal cells; however, it is unknown whether Ski is expressed and has a role in granulosa cells undergoing luteinization. Thus, the aim of the present study was to locate Ski protein in the rat ovary during luteinizationto predict the possible role of Ski. In order to examine the expression pattern of Ski protein along with the progress of luteinization, follicular growth was induced by administration of equine chorionic gonadtropin to immature female rats, and luteinization was induced by human chorionic gonadtropin treatment to mimic luteinizing hormone (LH) surge. While no Ski-positive granulosa cells were present in preovulatory follicle, Ski protein expression was induced in response to LH surge, and was maintained after the formation of the corpus luteum (CL). Though Ski protein is absent in granulosa cells of preovulatory follicle, its mRNA (c-Ski) was expressed and the level was unchanged even after LH surge. Taken together, these results demonstrated that Ski protein expression is induced in granulosa cells upon luteinization, and suggests that its expression is regulated post-transcriptionally.
Collapse
Affiliation(s)
- Hyun Kim
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan ; Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Dong Hun Kim
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Soo Bong Park
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Yeoung-Gyu Ko
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Sung-Woo Kim
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Yoon Jun Do
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Jae-Hong Park
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Boh-Suk Yang
- Korean Native Cattle Experiment Station, National Institute of Animal Science, RDA, Pyeongchang 232-952, Korea
| |
Collapse
|
17
|
Abstract
The transforming growth factor β (TGFβ) superfamily of signal transduction molecules plays crucial roles in the regulation of cell behavior. TGFβ regulates gene transcription through Smad proteins and signals via non-Smad pathways. The TGFβ pathway is strictly regulated, and perturbations lead to tumorigenesis. Several pathway components are known to be targeted for proteasomal degradation via ubiquitination by E3 ligases. Smurfs are well known negative regulators of TGFβ, which function as E3 ligases recruited by adaptors such as I-Smads. TGFβ signaling can also be enhanced by E3 ligases, such as Arkadia, that target repressors for degradation. It is becoming clear that E3 ligases often target multiple pathways, thereby acting as mediators of signaling cross-talk. Regulation via ubiquitination involves a complex network of E3 ligases, adaptor proteins, and deubiquitinating enzymes (DUBs), the last-mentioned acting by removing ubiquitin from its targets. Interestingly, also non-degradative ubiquitin modifications are known to play important roles in TGFβ signaling. Ubiquitin modifications thus play a key role in TGFβ signal transduction, and in this review we provide an overview of known players, focusing on recent advances.
Collapse
Affiliation(s)
- Miriam De Boeck
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|
18
|
Nishimori H, Ehata S, Suzuki HI, Katsuno Y, Miyazono K. Prostate cancer cells and bone stromal cells mutually interact with each other through bone morphogenetic protein-mediated signals. J Biol Chem 2012; 287:20037-46. [PMID: 22532569 DOI: 10.1074/jbc.m112.353094] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Functional interactions between cancer cells and the bone microenvironment contribute to the development of bone metastasis. Although the bone metastasis of prostate cancer is characterized by increased ossification, the molecular mechanisms involved in this process are not fully understood. Here, the roles of bone morphogenetic proteins (BMPs) in the interactions between prostate cancer cells and bone stromal cells were investigated. In human prostate cancer LNCaP cells, BMP-4 induced the production of Sonic hedgehog (SHH) through a Smad-dependent pathway. In mouse stromal MC3T3-E1 cells, SHH up-regulated the expression of activin receptor IIB (ActR-IIB) and Smad1, which in turn enhanced BMP-responsive reporter activities in these cells. The combined stimulation with BMP-4 and SHH of MC3T3-E1 cells cooperatively induced the expression of osteoblastic markers, including alkaline phosphatase, bone sialoprotein, collagen type II α1, and osteocalcin. When MC3T3-E1 cells and LNCaP cells were co-cultured, the osteoblastic differentiation of MC3T3-E1 cells, which was induced by BMP-4, was accelerated by SHH from LNCaP cells. Furthermore, LNCaP cells and BMP-4 cooperatively induced the production of growth factors, including fibroblast growth factor (FGF)-2 and epidermal growth factor (EGF) in MC3T3-E1 cells, and these may promote the proliferation of LNCaP cells. Taken together, our findings suggest that BMPs provide favorable circumstances for the survival of prostate cancer cells and the differentiation of bone stromal cells in the bone microenvironment, possibly leading to the osteoblastic metastasis of prostate cancer.
Collapse
Affiliation(s)
- Hikaru Nishimori
- Department of Molecular Pathology, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | |
Collapse
|
19
|
Kim H, Yamanouchi K, Matsuwaki T, Nishihara M. Induction of Ski protein expression upon luteinization in rat granulosa cells without a change in its mRNA expression. J Reprod Dev 2012; 58:254-9. [PMID: 22277932 DOI: 10.1262/jrd.11-118o] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Ski protein is implicated in the proliferation/differentiation of a variety of cells. We previously reported that the Ski protein is present in granulosa cells of atretic follicles, but not in preovulatory follicles, suggesting that Ski has a role in apoptosis of granulosa cells. However, granulosa cells cannot only undergo apoptosis but can alternatively differentiate into luteal cells. It is unknown whether Ski is expressed and has a role in granulosa cells undergoing luteinization. Thus, the aim of the present study was to determine the localization of the Ski protein in the rat ovary during luteinization to examine if Ski might play a role in this process. In order to examine the Ski protein expression during the progression of luteinization, follicular growth was induced in immature female rats by administration of equine chorionic gonadotropin, and luteinization was induced by human chorionic gonadotropin treatment to mimic the luteinizing hormone (LH) surge. While no Ski-positive granulosa cells were present in the preovulatory follicle, Ski protein expression was induced in response to the LH surge and was maintained after formation of the corpus luteum (CL). Although the Ski protein is absent from the granulosa cells of the preovulatory follicle, its mRNA (c-ski) was expressed, and the level of c-ski mRNA was unchanged even after the LH surge. The combined results demonstrated that Ski protein expression is induced in granulosa cells upon luteinization, and suggested that its expression is regulated posttranscriptionally.
Collapse
Affiliation(s)
- Hyun Kim
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | | | | | | |
Collapse
|
20
|
Kawata M, Koinuma D, Ogami T, Umezawa K, Iwata C, Watabe T, Miyazono K. TGF-β-induced epithelial-mesenchymal transition of A549 lung adenocarcinoma cells is enhanced by pro-inflammatory cytokines derived from RAW 264.7 macrophage cells. J Biochem 2011; 151:205-16. [PMID: 22161143 DOI: 10.1093/jb/mvr136] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cancer cells undergo epithelial-mesenchymal transition (EMT) during invasion and metastasis. Although transforming growth factor-β (TGF-β) and pro-inflammatory cytokines have been implicated in EMT, the underlying molecular mechanisms remain to be elucidated. Here, we studied the effects of proinflammatory cytokines derived from the mouse macrophage cell line RAW 264.7 on TGF-β-induced EMT in A549 lung cancer cells. Co-culture and treatment with conditioned medium of RAW 264.7 cells enhanced a subset of TGF-β-induced EMT phenotypes in A549 cells, including changes in cell morphology and induction of mesenchymal marker expression. These effects were increased by the treatment of RAW 264.7 cells with lipopolysaccharide, which also induced the expression of various proinflammatory cytokines, including TNF-α and IL-1β. The effects of conditioned medium of RAW 264.7 cells were partially inhibited by a TNF-α neutralizing antibody. Dehydroxy methyl epoxyquinomicin, a selective inhibitor of NFκB, partially inhibited the enhancement of fibronectin expression by TGF-β, TNF-α, and IL-1β, but not of N-cadherin expression. Effects of other pharmacological inhibitors also suggested complex regulatory mechanisms of the TGF-β-induced EMT phenotype by TNF-α stimulation. These findings provide direct evidence of the effects of RAW 264.7-derived TNF-α on TGF-β-induced EMT in A549 cells, which is transduced in part by NFκB signalling.
Collapse
Affiliation(s)
- Mikiko Kawata
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Sharma V, Antonacopoulou AG, Tanaka S, Panoutsopoulos AA, Bravou V, Kalofonos HP, Episkopou V. Enhancement of TGF-β signaling responses by the E3 ubiquitin ligase Arkadia provides tumor suppression in colorectal cancer. Cancer Res 2011; 71:6438-49. [PMID: 21998011 PMCID: PMC3194767 DOI: 10.1158/0008-5472.can-11-1645] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
TGF-β signaling provides tumor protection against colorectal cancer (CRC). Mechanisms that support its tumor-suppressive properties remain unclear. The ubiquitin ligase Arkadia/RNF111 enhances TGF-β signaling responses by targeting repressors of the pathway for degradation. The corepressors SnoN/Ski, critical substrates of Arkadia, complex with the activated TGF-β signaling effectors Smad2/3 (pSmad2/3) on the promoters of target genes and block their transcription. Arkadia degrades this complex including pSmad2/3 and unblocks the promoter. Here, we report that Arkadia is expressed highly in the mouse colonic epithelium. Heterozygous Akd(+/-) mice are normal but express less Arkadia. This leads to reduced expression of several TGF-β target genes, suggesting that normal levels of Arkadia are required for efficient signaling responses. Critically, Akd(+/-) mice exhibit increased susceptibility to azoxymethane/dextran sodium sulfate carcinogen-induced CRC, as they develop four-fold more tumors than wild-type mice. Akd(+/-) tumors also exhibit a more aggressive pathology, higher proliferation index, and reduced cytostasis. Therefore, Arkadia functions as a tumor suppressor whose peak expression is required to suppress CRC development and progression. The accumulation of nuclear SnoN and pSmad2, along with the downregulation of TGF-β target genes observed in Akd(+/-) colon and tumors, suggest that tumor-suppressing properties of Arkadia are mediated by its ability to derepress TGF-β signaling. Consistent with this likelihood, we identified mutations in primary colorectal tumors from human patients that reduce Arkadia function and are associated with the accumulation of nuclear SNON. Collectively, our findings reveal that Arkadia enhances TGF-β signaling responses and supports its tumor-suppressing properties in CRC.
Collapse
Affiliation(s)
- Vikas Sharma
- Department of Experimental Medicine, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
22
|
Bone morphogenetic protein-2 and -4 play tumor suppressive roles in human diffuse-type gastric carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2920-30. [PMID: 21996676 DOI: 10.1016/j.ajpath.2011.08.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 07/21/2011] [Accepted: 08/29/2011] [Indexed: 11/21/2022]
Abstract
A relationship exists between defects in bone morphogenetic protein (BMP) signaling and formation of hamartoma and adenoma in the gastric epithelium; however, the role of BMP signaling in the progression of diffuse-type gastric carcinoma remains unknown. We investigated whether BMP functions as a tumor suppressor in human diffuse-type gastric carcinoma using three different human diffuse-type gastric carcinoma cell lines (OCUM-12, HSC-39, and OCUM-2MLN). Overexpression of the dominant-negative form of BMP-2/4-specific type I receptor (ALK-3) in OCUM-12 and HSC-39 cells accelerated their growth in vivo. BMP-4 induced cell cycle arrest in these cells via p21 induction through the SMAD pathway. Moreover, overexpression of the constitutively active form of ALK-3 in HSC-39 and OCUM-2MLN cells suppressed the proliferation of these cells in vitro and in vivo. Our findings suggest that BMP-2 and BMP-4 function as potent tumor suppressors in diffuse-type gastric carcinoma.
Collapse
|
23
|
Mizutani A, Koinuma D, Tsutsumi S, Kamimura N, Morikawa M, Suzuki HI, Imamura T, Miyazono K, Aburatani H. Cell type-specific target selection by combinatorial binding of Smad2/3 proteins and hepatocyte nuclear factor 4alpha in HepG2 cells. J Biol Chem 2011; 286:29848-60. [PMID: 21646355 PMCID: PMC3191026 DOI: 10.1074/jbc.m110.217745] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Specific regulation of target genes by transforming growth factor-β (TGF-β) in a given cellular context is determined in part by transcription factors and cofactors that interact with the Smad complex. In this study, we determined Smad2 and Smad3 (Smad2/3) binding regions in the promoters of known genes in HepG2 hepatoblastoma cells, and we compared them with those in HaCaT epidermal keratinocytes to elucidate the mechanisms of cell type- and context-dependent regulation of transcription induced by TGF-β. Our results show that 81% of the Smad2/3 binding regions in HepG2 cells were not shared with those found in HaCaT cells. Hepatocyte nuclear factor 4α (HNF4α) is expressed in HepG2 cells but not in HaCaT cells, and the HNF4α-binding motif was identified as an enriched motif in the HepG2-specific Smad2/3 binding regions. Chromatin immunoprecipitation sequencing analysis of HNF4α binding regions under TGF-β stimulation revealed that 32.5% of the Smad2/3 binding regions overlapped HNF4α bindings. MIXL1 was identified as a new combinatorial target of HNF4α and Smad2/3, and both the HNF4α protein and its binding motif were required for the induction of MIXL1 by TGF-β in HepG2 cells. These findings generalize the importance of binding of HNF4α on Smad2/3 binding genomic regions for HepG2-specific regulation of transcription by TGF-β and suggest that certain transcription factors expressed in a cell type-specific manner play important roles in the transcription regulated by the TGF-β-Smad signaling pathway.
Collapse
Affiliation(s)
- Anna Mizutani
- From the Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033
| | - Daizo Koinuma
- From the Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033
| | - Shuichi Tsutsumi
- the Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, and
| | - Naoko Kamimura
- the Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, and
| | - Masato Morikawa
- From the Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033
| | - Hiroshi I. Suzuki
- From the Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033
| | - Takeshi Imamura
- the Division of Biochemistry, Cancer Institute of the Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Kohei Miyazono
- From the Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033
- To whom correspondence should be addressed. Tel.: 81-3-5841-3356; Fax: 81-3-5841-3354; E-mail:
| | - Hiroyuki Aburatani
- the Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, and
| |
Collapse
|
24
|
Koinuma D, Shinozaki M, Nagano Y, Ikushima H, Horiguchi K, Goto K, Chano T, Saitoh M, Imamura T, Miyazono K, Miyazawa K. RB1CC1 protein positively regulates transforming growth factor-beta signaling through the modulation of Arkadia E3 ubiquitin ligase activity. J Biol Chem 2011; 286:32502-12. [PMID: 21795712 PMCID: PMC3173165 DOI: 10.1074/jbc.m111.227561] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is controlled by a variety of regulators, of which Smad7, c-Ski, and SnoN play a pivotal role in its negative regulation. Arkadia is a RING-type E3 ubiquitin ligase that targets these negative regulators for degradation to enhance TGF-β signaling. In the present study we identified a candidate human tumor suppressor gene product RB1CC1/FIP200 as a novel positive regulator of TGF-β signaling that functions as a substrate-selective cofactor of Arkadia. Overexpression of RB1CC1 enhanced TGF-β signaling, and knockdown of endogenous RB1CC1 attenuated TGF-β-induced expression of target genes as well as TGF-β-induced cytostasis. RB1CC1 down-regulated the protein levels of c-Ski but not SnoN by enhancing the activity of Arkadia E3 ligase toward c-Ski. Substrate selectivity is primarily attributable to the physical interaction of RB1CC1 with substrates, suggesting its role as a scaffold protein. RB1CC1 thus appears to play a unique role as a modulator of TGF-β signaling by restricting substrate specificity of Arkadia.
Collapse
Affiliation(s)
- Daizo Koinuma
- Division of Biochemistry, The Cancer Institute of the Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Morikawa M, Koinuma D, Tsutsumi S, Vasilaki E, Kanki Y, Heldin CH, Aburatani H, Miyazono K. ChIP-seq reveals cell type-specific binding patterns of BMP-specific Smads and a novel binding motif. Nucleic Acids Res 2011; 39:8712-27. [PMID: 21764776 PMCID: PMC3203580 DOI: 10.1093/nar/gkr572] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Dysregulated bone morphogenetic protein (BMP) signaling in endothelial cells (ECs) and pulmonary arterial smooth muscle cells (PASMCs) are implicated in human genetic disorders. Here, we generated genome-wide maps of Smad1/5 binding sites in ECs and PASMCs. Smad1/5 preferentially bound to the region outside the promoter of known genes, and the binding was associated with target gene upregulation. Cell-selective Smad1/5 binding patterns appear to be determined mostly by cell-specific differences in baseline chromatin accessibility patterns. We identified, for the first time, a Smad1/5 binding motif in mammals, and termed GC-rich Smad binding element (GC-SBE). Several sequences in the identified GC-SBE motif had relatively weak affinity for Smad binding, and were enriched in cell type-specific Smad1/5 binding regions. We also found that both GC-SBE and the canonical SBE affect binding affinity for the Smad complex. Furthermore, we characterized EC-specific Smad1/5 target genes and found that several Notch signaling pathway-related genes were induced by BMP in ECs. Among them, a Notch ligand, JAG1 was regulated directly by Smad1/5, transactivating Notch signaling in the neighboring cells. These results provide insights into the molecular mechanism of BMP signaling and the pathogenesis of vascular lesions of certain genetic disorders, including hereditary hemorrhagic telangiectasia.
Collapse
MESH Headings
- Binding Sites
- Bone Morphogenetic Proteins/pharmacology
- Calcium-Binding Proteins/genetics
- Cell Line
- Chromatin Immunoprecipitation
- Enhancer Elements, Genetic
- Genome, Human
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Intercellular Signaling Peptides and Proteins/genetics
- Jagged-1 Protein
- Membrane Proteins/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Nucleotide Motifs
- Pulmonary Artery/cytology
- Receptors, Notch/metabolism
- Regulatory Elements, Transcriptional
- Sequence Analysis, DNA
- Serrate-Jagged Proteins
- Smad1 Protein/metabolism
- Smad5 Protein/metabolism
Collapse
Affiliation(s)
- Masato Morikawa
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Shuichi Tsutsumi
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Eleftheria Vasilaki
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Yasuharu Kanki
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Carl-Henrik Heldin
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Hiroyuki Aburatani
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan, Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden and Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
- *To whom correspondence should be addressed. Tel: +81 3 5841 3356; Fax: +81 3 5841 3354;
| |
Collapse
|
26
|
Bonnon C, Atanasoski S. c-Ski in health and disease. Cell Tissue Res 2011; 347:51-64. [DOI: 10.1007/s00441-011-1180-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/15/2011] [Indexed: 01/28/2023]
|
27
|
Hoshino Y, Katsuno Y, Ehata S, Miyazono K. Autocrine TGF-β protects breast cancer cells from apoptosis through reduction of BH3-only protein, Bim. J Biochem 2011; 149:55-65. [PMID: 20880961 DOI: 10.1093/jb/mvq114] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cancer cells undergo multi-step processes in obtaining the ability to metastasize, and are constantly exposed to signals that induce apoptosis. Acquisition of anti-apoptotic properties by cancer cells is important for metastasis, and recent studies suggest that transforming growth factor (TGF)-β promotes the survival of certain types of cancer cells. Here, we found that in highly metastatic breast cancer cells, JygMC(A), JygMC(B) and 4T1, TGF-β ligands were produced in autocrine fashion. Pharmacological inhibition of endogenous TGF-β signalling by a TGF-β type I receptor kinase inhibitor in serum-free conditions increased the expression of BH3-only protein, Bim (also known as Bcl2-like 11) in JygMC(A) and JygMC(B) cells, and caused apoptotic cell death. We also found that induction of Bim by TGF-β was not observed in Foxc1 knocked-down cancer cells. These findings suggest that TGF-β plays a crucial role in the regulation of survival of certain types of cancer cells through the TGF-β-Foxc1-Bim pathway, and that specific inhibitors of TGF-β signalling might be useful as apoptosis inducers in breast cancer cells.
Collapse
Affiliation(s)
- Yukari Hoshino
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
28
|
Inoue Y, Iemura SI, Natsume T, Miyazawa K, Imamura T. Suppression of p53 activity through the cooperative action of Ski and histone deacetylase SIRT1. J Biol Chem 2010; 286:6311-20. [PMID: 21149449 DOI: 10.1074/jbc.m110.177683] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ski was originally identified as an oncogene based on the fact that Ski overexpression transformed chicken and quail embryo fibroblasts. Consistent with these proposed oncogenic roles, Ski is overexpressed in various human tumors. However, whether and how Ski functions in mammalian tumorigenesis has not been fully investigated. Here, we show that Ski interacts with p53 and attenuates the biological functions of p53. Ski overexpression attenuated p53-dependent transactivation, whereas Ski knockdown enhanced the transcriptional activity of p53. Interestingly, Ski bound to the histone deacetylase SIRT1 and stabilized p53-SIRT1 interaction to promote p53 deacetylation, which subsequently decreased the DNA binding activity of p53. Consistent with the ability of Ski to inactivate p53, overexpressing Ski desensitized cells to genotoxic drugs and Nutlin-3, a small-molecule antagonist of Mdm2 that stabilizes p53 and activates the p53 pathway, whereas knocking down Ski increased the cellular sensitivity to these agents. These results indicate that Ski negatively regulates p53 and suggest that the p53-Ski-SIRT1 axis is an attractive target for cancer therapy.
Collapse
Affiliation(s)
- Yasumichi Inoue
- Division of Biochemistry, Cancer Institute of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | | | | | | | | |
Collapse
|
29
|
Abstract
Arkadia, also known as ring finger 111 (Rnf111), is an E3 ubiquitin ligase that amplifies transforming growth factor (TGF)-β family signalling through degradation of negative TGF-β signal regulators, i.e. Smad7, c-Ski and SnoN. Arkadia plays critical roles in early embryonic development through modulation of nodal signalling, as well as progression of tissue fibrosis and cancer through regulation of TGF-β signalling. Recent findings suggest that, similar to other ubiquitin ligases, including Smurf1 and 2, Arkadia regulates signalling pathways other than those of the TGF-β family. Arkadia interacts with the clathrin-adaptor 2 (AP2) complex and regulates endocytosis of certain cell surface receptors, leading to modulation of epidermal growth factor (EGF) and possibly other signalling pathways.
Collapse
Affiliation(s)
- Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | | |
Collapse
|
30
|
Mizutani A, Saitoh M, Imamura T, Miyazawa K, Miyazono K. Arkadia complexes with clathrin adaptor AP2 and regulates EGF signalling. J Biochem 2010; 148:733-41. [PMID: 20965945 DOI: 10.1093/jb/mvq127] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Arkadia is a positive regulator of transforming growth factor (TGF)-β signalling that induces ubiquitin-dependent degradation of several inhibitory proteins of TGF-β signalling through its C-terminal RING domain. We report here that, through yeast-two-hybrid screening for Arkadia-binding proteins, the µ2 subunit of clathrin-adaptor 2 (AP2) complex was identified as an interacting partner of Arkadia. Arkadia was located in both the nucleus and the cytosol in mammalian cells. The C-terminal YXXΦ-binding domain of the µ2 subunit associated with the N-terminal YALL motif of Arkadia. Arkadia ubiquitylated the µ2 subunit at Lys130. In addition, Arkadia interacted with the AP2 complex, and modified endocytosis of epidermal growth factor receptor (EGFR) induced by EGF. Arkadia thus appears to regulate EGF signalling by modulating endocytosis of EGFR through interaction with AP2 complex.
Collapse
Affiliation(s)
- Anna Mizutani
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|