1
|
Fu T, Park GC, Han JH, Shin JH, Park HH, Kim KS. MoRBP9 Encoding a Ran-Binding Protein Microtubule-Organizing Center Is Required for Asexual Reproduction and Infection in the Rice Blast Pathogen Magnaporthe oryzae. THE PLANT PATHOLOGY JOURNAL 2019; 35:564-574. [PMID: 31832037 PMCID: PMC6901248 DOI: 10.5423/ppj.oa.07.2019.0204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/18/2019] [Indexed: 06/10/2023]
Abstract
Like many fungal pathogens, the conidium and appressorium play key roles during polycyclic dissemination and infection of Magnaporthe oryzae. Ran-binding protein microtubule-organizing center (RanBPM) is a highly conserved nucleocytoplasmic protein. In animalia, RanBPM has been implicated in apoptosis, cell morphology, and transcription. However, the functional roles of RanBPM, encoded by MGG_00753 (named MoRBP9) in M. oryzae, have not been elucidated. Here, the deletion mutant ΔMorbp9 for MoRBP9 was generated via homologous recombination to investigate the functions of this gene. The ΔMorbp9 exhibited normal conidial germination and vegetative growth but dramatically reduced conidiation compared with the wild type, suggesting that MoRBP9 is involved in conidial production. ΔMorbp9 conidia failed to produce appressoria on hydrophobic surfaces, whereas ΔMorbp9 still developed aberrantly shaped appressorium-like structures at hyphal tips on the same surface, suggesting that MoRBP9 is involved in the morphology of appressorium-like structures from hyphal tips and is critical for development of appressorium from germ tubes. Taken together, our results indicated that MoRBP9 played a pleiotropic role in polycyclic dissemination and infection-related morphogenesis of M. oryzae.
Collapse
Affiliation(s)
| | | | | | | | | | - Kyoung Su Kim
- Corresponding author.: Phone) +82-33-250-6435, FAX) +82-33-259-5558, E-mail)
| |
Collapse
|
2
|
Salemi LM, Maitland MER, McTavish CJ, Schild-Poulter C. Cell signalling pathway regulation by RanBPM: molecular insights and disease implications. Open Biol 2018; 7:rsob.170081. [PMID: 28659384 PMCID: PMC5493780 DOI: 10.1098/rsob.170081] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 06/01/2017] [Indexed: 12/25/2022] Open
Abstract
RanBPM (Ran-binding protein M, also called RanBP9) is an evolutionarily conserved, ubiquitous protein which localizes to both nucleus and cytoplasm. RanBPM has been implicated in the regulation of a number of signalling pathways to regulate several cellular processes such as apoptosis, cell adhesion, migration as well as transcription, and plays a critical role during development. In addition, RanBPM has been shown to regulate pathways implicated in cancer and Alzheimer's disease, implying that RanBPM has important functions in both normal and pathological development. While its functions in these processes are still poorly understood, RanBPM has been identified as a component of a large complex, termed the CTLH (C-terminal to LisH) complex. The yeast homologue of this complex functions as an E3 ubiquitin ligase that targets enzymes of the gluconeogenesis pathway. While the CTLH complex E3 ubiquitin ligase activity and substrates still remain to be characterized, the high level of conservation between the complexes in yeast and mammals infers that the CTLH complex could also serve to promote the degradation of specific substrates through ubiquitination, therefore suggesting the possibility that RanBPM's various functions may be mediated through the activity of the CTLH complex.
Collapse
Affiliation(s)
- Louisa M Salemi
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| | - Matthew E R Maitland
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| | - Christina J McTavish
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| | - Caroline Schild-Poulter
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| |
Collapse
|
3
|
Tang WH, Zhuang XJ, Song SD, Wu H, Zhang Z, Yang YZ, Zhang HL, Mao JM, Liu DF, Zhao LM, Lin HC, Hong K, Ma LL, Qiao J, Qin W, Tang Y, Jiang H. Ran-binding protein M is associated with human spermatogenesis and oogenesis. Mol Med Rep 2017; 17:2257-2262. [PMID: 29207172 PMCID: PMC5783472 DOI: 10.3892/mmr.2017.8147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 10/06/2017] [Indexed: 12/02/2022] Open
Abstract
The aim of the present study was to explore the underlying mechanism and diagnostic potential of Ran-binding protein M (RanBPM) in human spermatogenesis and oogenesis. RanBPM expression in human testis and ovaries was analysed using polymerase chain reaction (PCR) and western blotting, and immunofluorescence was performed on testis and ovary tissue sections during different developmental stages of spermatogenesis and oogenesis using RanBPM antibodies. Interactions with a variety of functional proteins were also investigated. RanBPM mRNA and protein expression levels were determined by PCR and western blotting in the tissue sections. Results revealed that the mRNA expression levels were highest in the testis followed by the ovary. The RanBPM protein was predominantly localized in the nucleus of germ cells, and the expression levels were highest in pachytene spermatocytes and cells surrounding spermatids in testis tissue. In ovary cells, RanBPM was localized in the nucleus and cytoplasm. In conclusion, the results suggested that RanBPM may have multiple roles in the regulation of germ cell proliferation during human spermatogenesis and oogenesis. This research may provide a novel insight into the underlying molecular mechanism of RanBPM and may have implications for the clinical diagnosis and treatment of human infertility.
Collapse
Affiliation(s)
- Wen-Hao Tang
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Xin-Jie Zhuang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Shi-De Song
- Department of Urology, Rizhao People's Hospital, Rizhao, Shandong 276500, P.R. China
| | - Han Wu
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Zhe Zhang
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Yu-Zhuo Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hong-Liang Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Jia-Ming Mao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, P.R. China
| | - De-Feng Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Lian-Ming Zhao
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Hao-Cheng Lin
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Kai Hong
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Lu-Lin Ma
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Weibing Qin
- Key Laboratory of Male Reproduction and Genetics, National Health and Family Planning Commission, Family Planning Research Institute of Guangdong Province, Guangzhou, Guangdong 510600, P.R. China
| | - Yunge Tang
- Key Laboratory of Male Reproduction and Genetics, National Health and Family Planning Commission, Family Planning Research Institute of Guangdong Province, Guangzhou, Guangdong 510600, P.R. China
| | - Hui Jiang
- 1Department of Urology, The Third Hospital of Peking University, Beijing 100191, P.R. China
| |
Collapse
|
4
|
DNA methylation of the CDC2L1 gene promoter region decreases the expression of the CDK11p58 protein and reduces apoptosis in keloid fibroblasts. Arch Dermatol Res 2017; 310:107-115. [DOI: 10.1007/s00403-017-1801-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/25/2017] [Accepted: 11/22/2017] [Indexed: 01/05/2023]
|
5
|
Das S, Suresh B, Kim HH, Ramakrishna S. RanBPM: a potential therapeutic target for modulating diverse physiological disorders. Drug Discov Today 2017; 22:1816-1824. [PMID: 28847759 DOI: 10.1016/j.drudis.2017.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 06/26/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
The Ran-binding protein microtubule-organizing center (RanBPM) is a highly conserved nucleocytoplasmic protein involved in a variety of intracellular signaling pathways that control diverse cellular functions. RanBPM interacts with proteins that are linked to various diseases, including Alzheimer's disease (AD), schizophrenia (SCZ), and cancer. In this article, we define the characteristics of the scaffolding protein RanBPM and focus on its interaction partners in diverse physiological disorders, such as neurological diseases, fertility disorders, and cancer.
Collapse
Affiliation(s)
- Soumyadip Das
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Bharathi Suresh
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Hyongbum Henry Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, South Korea; Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
6
|
Abstract
Over the past two decades there has been a great deal of interest in the development of inhibitors of the cyclin-dependent kinases (CDKs). This attention initially stemmed from observations that different CDK isoforms have key roles in cancer cell proliferation through loss of regulation of the cell cycle, a hallmark feature of cancer. CDKs have now been shown to regulate other processes, particularly various aspects of transcription. The early non-selective CDK inhibitors exhibited considerable toxicity and proved to be insufficiently active in most cancers. The lack of patient selection biomarkers and an absence of understanding of the inhibitory profile required for efficacy hampered the development of these inhibitors. However, the advent of potent isoform-selective inhibitors with accompanying biomarkers has re-ignited interest. Palbociclib, a selective CDK4/6 inhibitor, is now approved for the treatment of ER+/HER2- advanced breast cancer. Current developments in the field include the identification of potent and selective inhibitors of the transcriptional CDKs; these include tool compounds that have allowed exploration of individual CDKs as cancer targets and the determination of their potential therapeutic windows. Biomarkers that allow the selection of patients likely to respond are now being discovered. Drug resistance has emerged as a major hurdle in the clinic for most protein kinase inhibitors and resistance mechanism are beginning to be identified for CDK inhibitors. This suggests that the selective inhibitors may be best used combined with standard of care or other molecularly targeted agents now in development rather than in isolation as monotherapies.
Collapse
Affiliation(s)
- Steven R Whittaker
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Aurélie Mallinger
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul Workman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul A Clarke
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom.
| |
Collapse
|
7
|
Zhou Y, Shen JK, Hornicek FJ, Kan Q, Duan Z. The emerging roles and therapeutic potential of cyclin-dependent kinase 11 (CDK11) in human cancer. Oncotarget 2016; 7:40846-40859. [PMID: 27049727 PMCID: PMC5130049 DOI: 10.18632/oncotarget.8519] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/28/2016] [Indexed: 12/17/2022] Open
Abstract
Overexpression and/or hyperactivation of cyclin-dependent kinases (CDKs) are common features of most cancer types. CDKs have been shown to play important roles in tumor cell proliferation and growth by controlling cell cycle, transcription, and RNA splicing. CDK4/6 inhibitor palbociclib has been recently approved by the FDA for the treatment of breast cancer. CDK11 is a serine/threonine protein kinase in the CDK family and recent studies have shown that CDK11 also plays critical roles in cancer cell growth and proliferation. A variety of genetic and epigenetic events may cause universal overexpression of CDK11 in human cancers. Inhibition of CDK11 has been shown to lead to cancer cell death and apoptosis. Significant evidence has suggested that CDK11 may be a novel and promising therapeutic target for the treatment of cancers. This review will focus on the emerging roles of CDK11 in human cancers, and provide a proof-of-principle for continued efforts toward targeting CDK11 for effective cancer treatment.
Collapse
Affiliation(s)
- Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jacson K. Shen
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Francis J. Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhenfeng Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| |
Collapse
|
8
|
Shin J, Sohn YC. Identification of Ran-binding protein M as a stanniocalcin 2 interacting protein and implications for androgen receptor activity. BMB Rep 2015; 47:643-8. [PMID: 25154718 PMCID: PMC4281344 DOI: 10.5483/bmbrep.2014.47.11.097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Indexed: 11/20/2022] Open
Abstract
Stanniocalcin (STC), a glycoprotein hormone originally discovered in fish, has been implicated in calcium and phosphate homeostasis. While fishes and mammals possess two STC homologs (STC1 and STC2), the physiological roles of STC2 are largely unknown compared with those of STC1. In this study, we identified Ran-binding protein M (RanBPM) as a novel binding partner of STC2 using yeast two-hybrid screening. The interaction between STC2 and RanBPM was confirmed in mammalian cells by immunoprecipitation. STC2 enhanced the RanBPM-mediated transactivation of liganded androgen receptor (AR), but not thyroid receptor β, glucocorticoid receptor, or estrogen receptor β. We also found that AR interacted with RanBPM in both the absence and presence of testosterone (T). Furthermore, we discovered that STC2 recruits RanBPM/AR complex in T-dependent manner. Taken together, our findings suggest that STC2 is a novel RanBPM-interacting protein that promotes AR transactivation.
Collapse
Affiliation(s)
- Jihye Shin
- Department of Marine Molecular Biotechnology, Gangneung-Wonju National University, Gangneung 210-702, Korea
| | - Young Chang Sohn
- Department of Marine Molecular Biotechnology, Gangneung-Wonju National University, Gangneung 210-702, Korea
| |
Collapse
|
9
|
Salemi LM, Loureiro SO, Schild-Poulter C. Characterization of RanBPM molecular determinants that control its subcellular localization. PLoS One 2015; 10:e0117655. [PMID: 25659156 PMCID: PMC4319831 DOI: 10.1371/journal.pone.0117655] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/30/2014] [Indexed: 12/14/2022] Open
Abstract
RanBPM/RanBP9 is a ubiquitous, nucleocytoplasmic protein that is part of an evolutionary conserved E3 ubiquitin ligase complex whose function and targets in mammals are still unknown. RanBPM itself has been implicated in various cellular processes that involve both nuclear and cytoplasmic functions. However, to date, little is known about how RanBPM subcellular localization is regulated. We have conducted a systematic analysis of RanBPM regions that control its subcellular localization using RanBPM shRNA cells to examine ectopic RanBPM mutant subcellular localization without interference from the endogenously expressed protein. We show that several domains and motifs regulate RanBPM nuclear and cytoplasmic localization. In particular, RanBPM comprises two motifs that can confer nuclear localization, one proline/glutamine-rich motif in the extreme N-terminus which has a dominant effect on RanBPM localization, and a second motif in the C-terminus which minimally contributes to RanBPM nuclear targeting. We also identified a nuclear export signal (NES) which mutation prevented RanBPM accumulation in the cytoplasm. Likewise, deletion of the central RanBPM conserved domains (SPRY and LisH/CTLH) resulted in the relocalization of RanBPM to the nucleus, suggesting that RanBPM cytoplasmic localization is also conferred by protein-protein interactions that promote its cytoplasmic retention. Indeed we found that in the cytoplasm, RanBPM partially colocalizes with microtubules and associates with α-tubulin. Finally, in the nucleus, a significant fraction of RanBPM is associated with chromatin. Altogether, these analyses reveal that RanBPM subcellular localization results from the combined effects of several elements that either confer direct transport through the nucleocytoplasmic transport machinery or regulate it indirectly, likely through interactions with other proteins and by intramolecular folding.
Collapse
Affiliation(s)
- Louisa M. Salemi
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sandra O. Loureiro
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Caroline Schild-Poulter
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
10
|
Salemi LM, Almawi AW, Lefebvre KJ, Schild-Poulter C. Aggresome formation is regulated by RanBPM through an interaction with HDAC6. Biol Open 2014; 3:418-30. [PMID: 24795145 PMCID: PMC4058076 DOI: 10.1242/bio.20147021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In conditions of proteasomal impairment, the build-up of damaged or misfolded proteins activates a cellular response leading to the recruitment of damaged proteins into perinuclear aggregates called aggresomes. Aggresome formation involves the retrograde transport of cargo proteins along the microtubule network and is dependent on the histone deacetylase HDAC6. Here we show that ionizing radiation (IR) promotes Ran-Binding Protein M (RanBPM) relocalization into discrete perinuclear foci where it co-localizes with aggresome components ubiquitin, dynein and HDAC6, suggesting that the RanBPM perinuclear clusters correspond to aggresomes. RanBPM was also recruited to aggresomes following treatment with the proteasome inhibitor MG132 and the DNA-damaging agent etoposide. Strikingly, aggresome formation by HDAC6 was markedly impaired in RanBPM shRNA cells, but was restored by re-expression of RanBPM. RanBPM was found to interact with HDAC6 and to inhibit its deacetylase activity. This interaction was abrogated by a RanBPM deletion of its LisH/CTLH domain, which also prevented aggresome formation, suggesting that RanBPM promotes aggresome formation through an association with HDAC6. Our results suggest that RanBPM regulates HDAC6 activity and is a central regulator of aggresome formation.
Collapse
Affiliation(s)
- Louisa M Salemi
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Ahmad W Almawi
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Karen J Lefebvre
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Caroline Schild-Poulter
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
11
|
Abstract
Ran-binding protein M (RanBPM) is a nucleocytoplasmic protein of yet unknown function. We have previously shown that RanBPM inhibits expression of the anti-apoptotic factor Bcl-2 and promotes apoptosis induced by DNA damage. Here we show that the effects of RanBPM on Bcl-2 expression occur through a regulation of the ERK signaling pathway. Transient and stable down-regulation of RanBPM stimulated ERK phosphorylation, leading to Bcl-2 up-regulation, while re-expression of RanBPM reversed these effects. RanBPM was found to inhibit MEK and ERK activation induced by ectopic expression of active RasV12. Activation of ERK by active c-Raf was also prevented by RanBPM. Expression of RanBPM correlated with a marked decrease in the protein levels of ectopically expressed active c-Raf and also affected the expression of endogenous c-Raf. RanBPM formed a complex with both active c-Raf, consisting of the C-terminal kinase domain, and endogenous c-Raf in mammalian cells. In addition, RanBPM was found to decrease the binding of Hsp90 to c-Raf. Finally, we show that loss of RanBPM expression confers increased cell proliferation and cell migration properties to HEK293 cells. Altogether, these findings establish RanBPM as a novel inhibitor of the ERK pathway through an interaction with the c-Raf complex and a regulation of c-Raf stability, and provide evidence that RanBPM loss of expression results in constitutive activation of the ERK pathway and promotes cellular events leading to cellular transformation and tumorigenesis.
Collapse
Affiliation(s)
| | - Caroline Schild-Poulter
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
12
|
Zhao XL, Campos AR. Insulin signalling in mushroom body neurons regulates feeding behaviour in Drosophila larvae. J Exp Biol 2012; 215:2696-702. [DOI: 10.1242/jeb.066969] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
SUMMARY
Whereas the pivotal role of insulin signalling in cell division, growth and differentiation is well documented, its role in the regulation of neuronal function and behaviour has recently become the focus of intense investigation. The simple organization of the Drosophila larval brain and the availability of genetic tools to impair the function of insulin receptor signalling in a spatially specific manner makes Drosophila an attractive model to investigate the role of the insulin pathway in specific behaviours. Here, we show that impairment of insulin signalling in the mushroom body neurons, a structure involved in associative learning, impairs feeding behaviour in the Drosophila larva.
Collapse
Affiliation(s)
- Xiao Li Zhao
- Department of Biology, McMaster University, Hamilton, ON, CanadaL8S 4K1
| | - Ana Regina Campos
- Department of Biology, McMaster University, Hamilton, ON, CanadaL8S 4K1
| |
Collapse
|
13
|
Suresh B, Ramakrishna S, Baek KH. Diverse roles of the scaffolding protein RanBPM. Drug Discov Today 2011; 17:379-87. [PMID: 22094242 DOI: 10.1016/j.drudis.2011.10.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 10/08/2011] [Accepted: 10/31/2011] [Indexed: 11/27/2022]
Abstract
Ran-binding protein microtubule-organizing center (RanBPM) appears to function as a scaffolding protein in several signal transduction pathways. RanBPM is a crucial component of multiprotein complexes that regulate the cellular function by modulating and/or assembling with a wide range of proteins in different intracellular regions and thereby mediate diverse cellular functions. This suggests a role for RanBPM as a scaffolding protein. In this article, we have summarized the diverse functions of RanBPM and its interacting partners that have been investigated to date. Also, we have categorized the role of RanBPM into four divisions: RanBPM as a modulator/protein stabilizer, regulator of transcription activity, cell cycle and neurological functions.
Collapse
Affiliation(s)
- Bharathi Suresh
- Department of Biomedical Science, CHA University, CHA General Hospital, Seoul 135-081, Republic of Korea
| | | | | |
Collapse
|
14
|
Wang L, Fu C, Cui Y, Xie Y, Yuan Y, Wang X, Chen H, Huang BR. The Ran-binding protein RanBPM can depress the NF-κB pathway by interacting with TRAF6. Mol Cell Biochem 2011; 359:83-94. [DOI: 10.1007/s11010-011-1002-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 07/19/2011] [Indexed: 12/11/2022]
|
15
|
Chang Y, Paramasivam M, Girgenti MJ, Walikonis RS, Bianchi E, LoTurco JJ. RanBPM regulates the progression of neuronal precursors through M-phase at the surface of the neocortical ventricular zone. Dev Neurobiol 2010; 70:1-15. [PMID: 19790105 DOI: 10.1002/dneu.20750] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Many of the mitoses that produce pyramidal neurons in neocortex occur at the dorsolateral surface of the lateral ventricles in the embryo. RanBPM was found in a yeast two-hybrid screen to potentially interact with citron kinase (CITK), a protein shown previously to localize to the surface of the lateral ventricles and to be essential to neurogenic mitoses. Similar to its localization in epithelia, RanBPM protein is concentrated at the adherens junctions in developing neocortex. The biochemical interaction between CITK and RanBPM was confirmed in coimmunoprecipitation and protein overlay experiments. To test for a functional role of RanPBM in vivo, we used in utero RNAi. RanBPM RNAi decreased the polarization of CITK to the ventricular surface, increased the number of cells in mitosis, and decreased the number of cells in cytokinesis. Finally, the effect of RanBPM knockdown on mitosis was reversed in embryos mutant for CITK. Together, these results indicate that RanBPM, potentially through interaction with CITK, plays a role in the progression of neocortical precursors through M-phase at the ventricular surface.
Collapse
Affiliation(s)
- Yoonjeung Chang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
16
|
Atabakhsh E, Bryce DM, Lefebvre KJ, Schild-Poulter C. RanBPM Has Proapoptotic Activities That Regulate Cell Death Pathways in Response to DNA Damage. Mol Cancer Res 2009; 7:1962-72. [DOI: 10.1158/1541-7786.mcr-09-0098] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Talbot JN, Skifter DA, Bianchi E, Monaghan DT, Toews ML, Murrin LC. Regulation of mu opioid receptor internalization by the scaffold protein RanBPM. Neurosci Lett 2009; 466:154-8. [PMID: 19788913 DOI: 10.1016/j.neulet.2009.09.048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 09/24/2009] [Accepted: 09/24/2009] [Indexed: 10/20/2022]
Abstract
Mu opioid receptors (MOP) are transducers of the pharmacological effects of many opioid drugs, including analgesia and tolerance/dependence. Previously, we observed increased MOP signaling during postnatal development that was not associated with increased MOP or G protein expression. A yeast two-hybrid screen of a human brain cDNA library using the MOP C-terminus as bait identified RanBPM as a potential MOP-interacting protein. RanBPM has been recognized as a multi-functional scaffold protein that interacts with a variety of signaling receptors/proteins. Co-immunoprecipitation studies in HEK293 cells indicated that RanBPM constitutively associates with MOP. Functionally, RanBPM had no effect on MOP-mediated inhibition of adenylyl cyclase, yet reduced agonist-induced endocytosis of MOP. Mechanistically, RanBPM interfered with beta arrestin2-GFP translocation stimulated by MOP but not alpha(1B)-adrenergic receptor activation, indicating selectivity of action. Our findings suggest that RanBPM is a novel MOP-interacting protein that negatively regulates receptor internalization without altering MOP signaling through adenylyl cyclase.
Collapse
Affiliation(s)
- Jeffery N Talbot
- Department of Pharmacology and Experimental Neuroscience, 985800 Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | | | | | | | | | | |
Collapse
|
18
|
Neuronal expression of muskelin in the rodent central nervous system. BMC Neurosci 2007; 8:28. [PMID: 17474996 PMCID: PMC1876237 DOI: 10.1186/1471-2202-8-28] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 05/02/2007] [Indexed: 12/25/2022] Open
Abstract
Background The kelch repeat protein muskelin mediates cytoskeletal responses to the extracellular matrix protein thrombospondin 1, (TSP1), that is known to promote synaptogenesis in the central nervous system (CNS). Muskelin displays intracellular localization and affects cytoskeletal organization in adherent cells. Muskelin is expressed in adult brain and has been reported to bind the Cdk5 activator p39, which also facilitates the formation of functional synapses. Since little is known about muskelin in neuronal tissues, we here analysed the tissue distribution of muskelin in rodent brain and analysed its subcellular localization using cultured neurons from multiple life stages. Results Our data show that muskelin transcripts and polypeptides are expressed throughout the central nervous system with significantly high levels in hippocampus and cerebellum, a finding that resembles the tissue distribution of p39. At the subcellular level, muskelin is found in the soma, in neurite projections and the nucleus with a punctate distribution in both axons and dendrites. Immunostaining and synaptosome preparations identify partial localization of muskelin at synaptic sites. Differential centrifugation further reveals muskelin in membrane-enriched, rather than cytosolic fractions. Conclusion Our results suggest that muskelin represents a multifunctional protein associated with membranes and/or large protein complexes in most neurons of the central nervous system. These data are in conclusion with distinct roles of muskelin's functional interaction partners.
Collapse
|
19
|
Kobayashi N, Yang J, Ueda A, Suzuki T, Tomaru K, Takeno M, Okuda K, Ishigatsubo Y. RanBPM, Muskelin, p48EMLP, p44CTLH, and the armadillo-repeat proteins ARMC8alpha and ARMC8beta are components of the CTLH complex. Gene 2007; 396:236-47. [PMID: 17467196 DOI: 10.1016/j.gene.2007.02.032] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 02/13/2007] [Accepted: 02/23/2007] [Indexed: 11/17/2022]
Abstract
Ran-binding protein in microtubule organising centre (RanBPM) was originally isolated as a protein that binds to the small GTPase Ran. Recently our group and other groups reported that RanBPM was associated with several proteins and composed a large protein complex. Here, we used tandem MS with an antibody against RanBPM to purify this complex from a soluble extract of HEK293 cells: we identified Muskelin, p48EMLP, p44CTLH, and the novel armadillo-repeat proteins ARMC8alpha and ARMC8beta as components. In RanBPM, Muskelin, p48EMLP, and p44CTLH we found LisH/CTLH motifs, which are present in proteins involved in microtubule dynamics, cell migration, nucleokinesis, and chromosome segregation. We renamed the 20S large protein complex the CTLH complex. The N-terminal 364 amino acids of ARMC8alpha and ARMC8beta were completely conserved, suggesting that these proteins are probably alternatively spliced products from the same gene. We confirmed the in vivo association of each component by co-immunoprecipitation assays with Cos-7 cells in which these components were exogenously overexpressed. A pull-down assay with bacterially-expressed Twa1 revealed binding of each in vitro-translated component to Twa1. Finally, we confirmed the cellular localization of these proteins. Taken together, our results reveal that RanBPM, ARMC8alpha, ARMC8beta, Muskelin, p48EMLP, and p44CTLH form complexes in cells.
Collapse
Affiliation(s)
- Nobuaki Kobayashi
- Department of Internal Medicine and Clinical Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama-City 236-0004, Japan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Yuan Y, Fu C, Chen H, Wang X, Deng W, Huang BR. The Ran binding protein RanBPM interacts with TrkA receptor. Neurosci Lett 2006; 407:26-31. [PMID: 16959415 DOI: 10.1016/j.neulet.2006.06.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 06/20/2006] [Accepted: 06/28/2006] [Indexed: 10/24/2022]
Abstract
RanBPM as a novel binding protein can interact with neurotrophin receptor p75NTR and tyrosine kinase receptor Met which has a similar tyrosine kinase structure as receptor TrkA has. Whether RanBPM interacts with neurotrophin receptor TrkA has not been established to date. In this study, using yeast two-hybrid system, it was identified that RanBPM bound to the intracellular domain (ICD) of neurotrophin receptor TrkA through its SPRY motif. We confirmed the formation of complexes between RanBPM and TrkA by co-immunoprecipitation studies and GST pull-down assays. The region of TrkA interacted with the SPRY domain of RanBPM was located in its tyrosine kinase domain. Furthermore, coimmunoprecipitaiton revealed endogenous RanBPM and receptors TrkA did interact in several mammalian cell lines. It was found that the overexpression of RanBPM could inhibit NGF-induced increase of nuclear factor of activated T cells (NFAT) dependent luciferase expression through its interaction with receptor TrkA, and NFAT transcriptional activity plays an important role in neuronal signal transduction. These data suggested that RanBPM could participate in neurotrophin-mediated gene transcription and expression by its binding to TrkA.
Collapse
Affiliation(s)
- Yuhe Yuan
- National Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | | | | | | | | | | |
Collapse
|
21
|
Xie J, Sun M, Guo L, Liu W, Jiang J, Chen X, Zhou L, Gu J. Human Dectin-1 isoform E is a cytoplasmic protein and interacts with RanBPM. Biochem Biophys Res Commun 2006; 347:1067-73. [PMID: 16870151 DOI: 10.1016/j.bbrc.2006.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 07/01/2006] [Indexed: 10/24/2022]
Abstract
Human Dectin-1, a type II transmembrane receptor, is alternatively spliced, generating eight isoforms. Of these isoforms, the isoform E (hDectin-1E) is structurally unique, containing a complete C-type lectin-like domain as well as an ITAM-like sequence. So far, little is known about its function. In the present study, we demonstrated that hDectin-1E was not secreted and it mainly resided in the cytoplasm. Using yeast two-hybrid screening, we identified a Ran-binding protein, RanBPM, as an interacting partner of hDectin-1E. GST pull-down assays showed that RanBPM interacted directly with hDectin-1E and the region containing SPRY domain was sufficient for the interaction. The binding of hDectin-1E and RanBPM was further confirmed in vivo by co-immunoprecipitation assay and confocal microscopic analysis. Taken together, our data provide a clue to the understanding of the function about hDectin-1E.
Collapse
Affiliation(s)
- Jianhui Xie
- State Key Laboratory of Genetic Engineering and Gene Research Center, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Wu Y, Sun X, Kaczmarek E, Dwyer K, Bianchi E, Usheva A, Robson S. RanBPM associates with CD39 and modulates ecto-nucleotidase activity. Biochem J 2006; 396:23-30. [PMID: 16478441 PMCID: PMC1449986 DOI: 10.1042/bj20051568] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Revised: 02/07/2006] [Accepted: 02/14/2006] [Indexed: 01/10/2023]
Abstract
CD39/ecto-NTPDase 1 (nucleoside triphosphate diphosphohydrolase 1) is an ecto-nucleotidase that influences P2 receptor activation to regulate vascular and immune cell adhesion and signalling events pivotal in inflammation. Whether CD39 interacts with other membrane or cytoplasmic proteins has not been established to date. Using the yeast two-hybrid system, we note that the N-terminus of CD39 binds to RanBPM (Ran binding protein M; also known as RanBP9), a multi-adaptor scaffolding membrane protein originally characterized as a binding protein for the small GTPase Ran. We confirm formation of complexes between CD39 and RanBPM in transfected mammalian cells by co-immunoprecipitation studies. Endogenous CD39 and RanBPM are also found to be co-expressed and abundant in cell membranes of B-lymphocytes. NTPDase activity of recombinant CD39, but not of N-terminus-deleted-CD39 mutant, is substantially diminished by RanBPM co-expression in COS-7 cells. The conserved SPRY [repeats in splA and RyR (ryanodine receptor)] moiety of RanBPM is insufficient alone for complete physical and functional interactions with CD39. We conclude that CD39 associations with RanBPM have the potential to regulate NTPDase catalytic activity. This intermolecular interaction may have important implications for the regulation of extracellular nucleotide-mediated signalling.
Collapse
Key Words
- cd39
- ecto-nucleotidase
- nucleoside triphosphate diphosphohydrolase 1 (ntpdase1)
- ranbpm
- spry domain
- yeast two-hybrid system
- acr, apyrase conserved region
- c-met, an rptk (receptor protein-tyrosine kinase) for hgf (hepatocyte growth factor)
- dc, dendritic cells
- dmem, dulbecco's modified eagle's medium
- ebv, epstein–barr virus
- fbs, fetal bovine serum
- ntpdase1, nucleoside triphosphate diphosphohydrolase 1
- p2y1, p2 (purinergic type-2)-receptor 1
- ranbpm, ran binding protein m
- ryr, ryanodine receptor
- sd, synthetic dropout
- spry, repeats in spla and ryr
Collapse
Affiliation(s)
- Yan Wu
- *Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | - Xiaofeng Sun
- *Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | - Elzbieta Kaczmarek
- *Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | - Karen M. Dwyer
- *Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | - Elisabetta Bianchi
- †Laboratory of Immunoregulation, Department of Immunology, Institut Pasteur, 25 rue du Dr. Roux, 75724 Paris, France
| | - Anny Usheva
- ‡Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | - Simon C. Robson
- *Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, U.S.A
| |
Collapse
|
23
|
Feng Y, Ariza M, Goulet AC, Shi J, Nelson M. Death-signal-induced relocalization of cyclin-dependent kinase 11 to mitochondria. Biochem J 2005; 392:65-73. [PMID: 16004605 PMCID: PMC1317665 DOI: 10.1042/bj20050195] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 07/07/2005] [Accepted: 07/11/2005] [Indexed: 12/26/2022]
Abstract
Fas receptor-Fas ligand interaction appears to be important in carcinogenesis, tumour outgrowth and metastasis. Emerging evidence suggests that CDK11 (cyclin-dependent kinase 11) plays a role in apoptosis and melanoma development. Here, we show that CDK11p110 protein kinase was cleaved after induction of apoptosis by Fas. The N-terminal portion of CDK11p110, CDK11p60, was translocated from the nucleus to the mitochondria. The targeting of CDK11p60 to mitochondria occurred as early as 12 h after treatment. Overexpression of EGFP (enhanced green fluorescent protein)-tagged CDK11p60 could partially break down the mitochondrial membrane potential, induce cytochrome c release and promote apoptosis. Reduction of endogenous CDK11p110 protein levels with siRNA (small interfering RNA) resulted in the suppression of both cytochrome c release and apoptosis. In addition, subcellular fractionation studies of Fas-mediated apoptosis demonstrated that CDK11p60 was associated with the mitochondrial import motor, mitochondrial heat shock protein 70. Taken together, our data suggest that CDK11p60 can contribute to apoptosis by direct signalling at the mitochondria, thereby amplifying Fas-induced apoptosis in melanoma cells.
Collapse
Key Words
- apoptosis
- cyclin-dependent kinase 11 (cdk11)
- mitochondria
- mitochondrial heat-shock protein (hsp70)
- 7-aad, 7-aminoactinomycin
- cdk11, cyclin-dependent kinase 11
- dapi, 4,6-diamidino-2-phenylindole
- egfp, enhanced green fluorescent protein
- eif3f, eukaryotic initiation factor 3f
- hsp60/90, heat-shock proteins 60 and 90 respectively
- mt-hsp70, mitochondrial hsp70
- parp, poly(adp-ribose) polymerase
- rt-pcr, reverse transcriptase-pcr
- sirna, small interfering rna
- tim, translocase of the inner membrane
- tom, translocase of the outer membrane
Collapse
Affiliation(s)
- Yongmei Feng
- Department of Pathology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85724, U.S.A
| | - Maria E. Ariza
- Department of Pathology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85724, U.S.A
| | - Anne-Christine Goulet
- Department of Pathology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85724, U.S.A
| | - Jiaqi Shi
- Department of Pathology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85724, U.S.A
| | - Mark A. Nelson
- Department of Pathology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85724, U.S.A
| |
Collapse
|
24
|
Hafizi S, Gustafsson A, Stenhoff J, Dahlbäck B. The Ran binding protein RanBPM interacts with Axl and Sky receptor tyrosine kinases. Int J Biochem Cell Biol 2005; 37:2344-56. [PMID: 15964779 DOI: 10.1016/j.biocel.2005.05.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Indexed: 12/14/2022]
Abstract
Axl belongs to a particular subfamily of transmembrane receptor tyrosine kinases, the biological ligand for which is the growth/survival factor Gas6. However, little is known about the molecular mechanisms for Axl activation and signal transduction. We have previously identified a novel interaction between the intracellular domain of Axl and Ran binding protein in microtubule organising centre (RanBPM). In the present study, we investigated further the nature of the RanBPM interaction with Axl. A wide distribution of RanBPM mRNA expression in human tissues and various human cancer cell lines was detected. The strength of interaction of both proteins in yeast was comparable to that with the other Axl-binding proteins phosphatidylinositol 3-kinase and Grb2. A truncated version of RanBPM with the SPRY-LisH domain region omitted failed to interact with Axl in yeast. RanBPM was also found to interact in yeast with the Axl homologue, Sky/Tyro3. The interaction between Axl intracellular domain and RanBPM was reproduced in coimmunoprecipitation experiments in both cell-free and mammalian cell systems. Furthermore, coimmunoprecipitation revealed endogenous Axl and RanBPM to interact in several mammalian cell lines in a constitutive manner. Stimulation of COS cells with Gas6 caused increased Axl tyrosine phosphorylation although appeared not to influence the RanBPM-Axl association. In conclusion, we have identified and characterised a novel interaction between RanBPM and the related receptor tyrosine kinases, Axl and Sky. This novel insight into the signalling interactions of Axl and Sky may shed further light on their suspected roles in tumourigenesis, inflammation as well as other cell proliferative diseases.
Collapse
Affiliation(s)
- Sassan Hafizi
- Department of Laboratory Medicine, Section for Clinical Chemistry, Lund University, Wallenberg Laboratory, University Hospital Malmö, SE-205 02 Malmö, Sweden.
| | | | | | | |
Collapse
|
25
|
Kotoshiba S, Kamura T, Hara T, Ishida N, Nakayama KI. Molecular Dissection of the Interaction between p27 and Kip1 Ubiquitylation-promoting Complex, the Ubiquitin Ligase That Regulates Proteolysis of p27 in G1 Phase. J Biol Chem 2005; 280:17694-700. [PMID: 15746103 DOI: 10.1074/jbc.m500866200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The cyclin-dependent kinase (CDK) inhibitor p27 is degraded at the G(0)-G(1) transition of the cell cycle by the ubiquitin-proteasome pathway in a Skp2-independent manner. We recently identified a novel ubiquitin ligase, KPC (Kip1 ubiquitylation-promoting complex), consisting of KPC1 and KPC2, which regulates the ubiquitin-dependent degradation of p27 at G(1) phase. We have now investigated the structural requirements for the interactions of KPC1 with KPC2 and p27. The NH(2)-terminal region of KPC1 was found to be responsible for binding to KPC2 and to p27. KPC1 mutants that lack this region failed to mediate polyubiquitylation of p27 in vitro and expression of one such mutant delayed p27 degradation in vivo. We also generated a series of deletion mutants of p27 and found that KPC failed to polyubiquitylate a p27 mutant that lacks the CDK inhibitory domain. Interestingly, the cyclin E.CDK2 complex prevented both the interaction of KPC with p27 as well as KPC-mediated polyubiquitylation of p27. A complex of cyclin E with a kinase-negative mutant of CDK2 also exhibited these inhibitory effects, suggesting that cyclin E.CDK2 competes with KPC1 for access to the CDK inhibitory domain of p27. These results suggest that free p27 is recognized by the NH(2)-terminal region of KPC1, which also associates with KPC2, and that p27 is then polyubiquitylated by the COOH-terminal RING-finger domain of KPC1.
Collapse
Affiliation(s)
- Shuhei Kotoshiba
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582
| | | | | | | | | |
Collapse
|
26
|
Wang D, Li Z, Messing EM, Wu G. The SPRY domain-containing SOCS box protein 1 (SSB-1) interacts with MET and enhances the hepatocyte growth factor-induced Erk-Elk-1-serum response element pathway. J Biol Chem 2005; 280:16393-401. [PMID: 15713673 DOI: 10.1074/jbc.m413897200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The suppressor of cytokine signaling (SOCS) protein family includes a SPRY (repeats in splA and RyR) domain-containing SOCS box protein (SSB) subfamily, which consists of four members, SSB-1, SSB-2, SSB-3, and SSB-4. These proteins contain a central SPRY domain and a C-terminal SOCS box. Although some of the SOCS protein subfamilies function as adaptors for a large family of ubiquitin-protein isopeptide ligases to regulate certain signaling pathways, the function of the SSB subfamily remains to be determined. In our previous studies, we have found that two SPRY domain-containing proteins, RanBP9 and RanBP10, interact with MET through the SPRY domain. In the present study, we explored the function of SSB proteins in the regulation of the hepatocyte growth factor (HGF)-MET signaling. Our results showed that all four SSB proteins also interacted with the MET. The MET interaction with SSB-1 was further investigated. We demonstrated that SSB-1 bound to MET tyrosine kinase domain through its SPRY domain. MET interacted with SSB-1 in both the absence and the presence of HGF, but HGF treatment resulted in the recruitment of more SSB-1 by MET. We showed that overexpression of SSB-1 but not other SSB proteins enhanced the HGF-induced serum response element (SRE)-luciferase activity. Overexpression of SSB-1 exhibited no effect on the basal level or epidermal growth factor-induced SRE-luciferase activity. SSB-1 also enhanced HGF-induced Erk phosphorylation. Suppression of SSB-1 by the RNA interference method down-regulated HGF-induced SRE-luciferase activity and decreased Elk-1 activation. These results suggest that SSB-1 may play an important role in enhancing the HGF-induced Erk-Elk-1-SRE pathway. Furthermore, we demonstrated that in response to HGF stimulation, the SSB-1 protein became phosphorylated at tyrosine residue 31. The phosphorylated SSB-1 protein bound to p120Ras-GTPase-activating protein (GAP) but did not promote the degradation of p120RasGAP, indicating that enhanced HGF-MET signaling by overexpression of SSB-1 was not dependent on p120RasGAP degradation.
Collapse
Affiliation(s)
- Dakun Wang
- Department of Urology, Department of Pathology and Laboratory Medicine, and The James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|