1
|
Mattoo S, Arora M, Sharma P, Pore SK. Targeting mammalian N-end rule pathway for cancer therapy. Biochem Pharmacol 2025; 231:116684. [PMID: 39613115 DOI: 10.1016/j.bcp.2024.116684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2024] [Revised: 11/12/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Regulated protein degradation plays a crucial role in maintaining proteostasis along with protein refolding and compartmentalisation which collectively control biological functions. The N-end rule pathway is a major ubiquitin-dependent protein degradation system. The short-lived protein substrates containing destabilizing amino acid residues (N-degrons) are recognized by E3 ubiquitin ligases containing UBR box domains (N-recognin) for degradation. The dysregulated pathway fails to maintain the metabolic stability of the substrate proteins which leads to diseases. The mammalian substrates of this pathway are involved in many hallmarks of cancer such as resisting cell death, evading growth suppression, chromosomal instability, angiogenesis, and deregulation of cellular metabolism. Besides, mutations in E3 N-recognin have been detected in human cancers. In this review, we discuss the mammalian N-end rule pathway components, functions, and mechanism of degradation of substrates, and their implications in cancer pathogenesis. We also discuss the impact of pharmacological and genetic inhibition of this pathway component on cancer cells and chemoresistance. We further highlight how this pathway can be manipulated for selective protein degradation; for instance, using PROTAC technique. The challenges and future perspectives to utilize this pathway as a drug target for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Shria Mattoo
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India
| | - Muskaan Arora
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India
| | - Priyanka Sharma
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Noida 201311, India
| | - Subrata Kumar Pore
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India.
| |
Collapse
|
2
|
Signaling Pathways Regulated by UBR Box-Containing E3 Ligases. Int J Mol Sci 2021; 22:ijms22158323. [PMID: 34361089 PMCID: PMC8346999 DOI: 10.3390/ijms22158323] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022] Open
Abstract
UBR box E3 ligases, also called N-recognins, are integral components of the N-degron pathway. Representative N-recognins include UBR1, UBR2, UBR4, and UBR5, and they bind destabilizing N-terminal residues, termed N-degrons. Understanding the molecular bases of their substrate recognition and the biological impact of the clearance of their substrates on cellular signaling pathways can provide valuable insights into the regulation of these pathways. This review provides an overview of the current knowledge of the binding mechanism of UBR box N-recognin/N-degron interactions and their roles in signaling pathways linked to G-protein-coupled receptors, apoptosis, mitochondrial quality control, inflammation, and DNA damage. The targeting of these UBR box N-recognins can provide potential therapies to treat diseases such as cancer and neurodegenerative diseases.
Collapse
|
3
|
Chen KY, Tsai TY, Chang CF, Ou JC, Tsai YR, Ma HP, Chiu WT, Tsai SH, Liao KH, Lin JW, Lin CM, Wu JCC, Chiang YH. Worsening of Dizziness Impairment Is Associated with Bone Marrow Kinase on Chromosome X Level in Patients after Mild Traumatic Brain Injury. J Neurotrauma 2021; 38:1445-1449. [PMID: 25747875 DOI: 10.1089/neu.2014.3691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022] Open
Abstract
Over 2 million people suffer from mild traumatic brain injury (mTBI) each year. Predicting symptoms of mTBI and the characterization of those symptoms has been challenging. Biomarkers that correlate clinical symptoms to disease outcome are desired to improve understanding of the disease and optimize patient care. Bone marrow kinase on chromosome X (BMX), a member of the TEC family of nonreceptor tyrosine kinases, is up-regulated after traumatic neural injury in a rat model of mTBI. The aim of this investigation was to determine whether BMX serum concentrations can effectively be used to predict outcomes after mTBI in a clinical setting. A total of 63 patients with mTBI (Glasgow Coma Score [GCS] between 13 and 15) were included. Blood samples taken at the time of hospital admission were analyzed for BMX. Data collected included demographic and clinical variables. Outcomes were assessed using the Dizziness Handicap Inventory (DHI) questionnaire at baseline and 6 weeks postinjury. The participant was asssigned to the case group if the subject's complaints of dizziness became worse at the sixth week assessment; otherwise, the participant was assigned to the control group. A receiver operating characteristic curve was constructed to explore BMX level. Significant associations were found between serum levels of BMX and dizziness. Areas under the curve for prediction of change in DHI postinjury were 0.76 for total score, 0.69 for physical score, 0.65 for emotional score, and 0.66 for functional score. Specificities were between 0.69 and 0.77 for total score and emotional score, respectively. Therefore, BMX demonstrates potential as a candidate serum biomarker of exacerbating dizziness post-mTBI.
Collapse
Affiliation(s)
- Kai-Yun Chen
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - Tung-Yao Tsai
- Department of Emergency Medicine, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Fu Chang
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ju-Chi Ou
- Department of Emergency Medicine, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yan-Rou Tsai
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - Hon-Ping Ma
- Department of Emergency Medicine, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wen-Ta Chiu
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shin-Han Tsai
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,Department of Emergency Medicine, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Hsing Liao
- Department of Neurosurgery, Wan Fang Hospital, Taipei, Taiwan
| | - Jia-Wei Lin
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chien-Min Lin
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - John Chung-Che Wu
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yung-Hsiao Chiang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Translational Research Laboratory, Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
4
|
Regulating Apoptosis by Degradation: The N-End Rule-Mediated Regulation of Apoptotic Proteolytic Fragments in Mammalian Cells. Int J Mol Sci 2018; 19:ijms19113414. [PMID: 30384441 PMCID: PMC6274719 DOI: 10.3390/ijms19113414] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/13/2018] [Revised: 10/24/2018] [Accepted: 10/27/2018] [Indexed: 12/13/2022] Open
Abstract
A pivotal hallmark of some cancer cells is the evasion of apoptotic cell death. Importantly, the initiation of apoptosis often results in the activation of caspases, which, in turn, culminates in the generation of proteolytically-activated protein fragments with potentially new or altered roles. Recent investigations have revealed that the activity of a significant number of the protease-generated, activated, pro-apoptotic protein fragments can be curbed via their selective degradation by the N-end rule degradation pathways. Of note, previous work revealed that several proteolytically-generated, pro-apoptotic fragments are unstable in cells, as their destabilizing N-termini target them for proteasomal degradation via the N-end rule degradation pathways. Remarkably, previous studies also showed that the proteolytically-generated anti-apoptotic Lyn kinase protein fragment is targeted for degradation by the UBR1/UBR2 E3 ubiquitin ligases of the N-end rule pathway in chronic myeloid leukemia cells. Crucially, the degradation of cleaved fragment of Lyn by the N-end rule counters imatinib resistance in these cells, implicating a possible linkage between the N-end rule degradation pathway and imatinib resistance. Herein, we highlight recent studies on the role of the N-end rule proteolytic pathways in regulating apoptosis in mammalian cells, and also discuss some possible future directions with respect to apoptotic proteolysis signaling.
Collapse
|
5
|
Li Y, Cui N, Zheng PS, Yang WT. BMX/Etk promotes cell proliferation and tumorigenicity of cervical cancer cells through PI3K/AKT/mTOR and STAT3 pathways. Oncotarget 2018; 8:49238-49252. [PMID: 28514765 PMCID: PMC5564764 DOI: 10.18632/oncotarget.17493] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2017] [Accepted: 04/17/2017] [Indexed: 11/25/2022] Open
Abstract
Bone marrow X-linked kinase (BMX, also known as Etk) has been reported to be involved in cell proliferation, differentiation, apoptosis, migration and invasion in several types of tumors, but its role in cervical carcinoma remains poorly understood. In this study, we showed that BMX expression exhibits a gradually increasing trend from normal cervical tissue to cervical cancer in situ and then to invasive cervical cancer tissue. Through BMX-IN-1, a potent and irreversible BMX kinase inhibitor, inhibited the expression of BMX, the cell proliferation was significantly decreased. Knockdown of BMX in HeLa and SiHa cervical cancer cell lines using two different silencing technologies, TALEN and shRNA, inhibited cell growth in vitro and suppressed xenograft tumor formation in vivo, whereas overexpression of BMX in the cell line C-33A significantly increased cell proliferation. Furthermore, a mechanism study showed that silencing BMX blocked cell cycle transit from G0/G1 to S or G2/M phase, and knockdown of BMX inhibited the expression of p-AKT and p-STAT3. These results suggested that BMX can promote cell proliferation through PI3K/AKT/mTOR and STAT3 signaling pathways in cervical cancer cells.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Reproductive Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Nan Cui
- Department of Reproductive Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Peng-Sheng Zheng
- Department of Reproductive Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.,Section of Cancer Stem Cell Research, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of The People's Republic of China, Xi'an, People's Republic of China
| | - Wen-Ting Yang
- Department of Reproductive Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
6
|
Wang Y, Xia J, Fang Z, Li F, Li D, Wang Z, Feng Y, Zhang J, Chen H, Ji H, Liu H. A novel BMX variant promotes tumor cell growth and migration in lung adenocarcinoma. Oncotarget 2018; 8:33405-33415. [PMID: 28422715 PMCID: PMC5464877 DOI: 10.18632/oncotarget.16796] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2017] [Accepted: 03/22/2017] [Indexed: 11/25/2022] Open
Abstract
The non-receptor tyrosine kinase BMX has been reported in several solid tumors. However, the alternative splicing of BMX and its clinical relevance in lung cancer remain to be elucidated. Exon1.0 array was used to identify a novel alternative splicing of BMX, BMXΔN, which was confirmed by rapid amplification of cDNA ends and reverse transcription-polymerase chain reaction. BMXΔN, resulting from exon skipping with excluding exon 1 to exon 8 of BMX gene, was found in 12% human lung adenocarcinoma specimens. BMXΔN is not found in paired pathologically normal lungs and positively correlated with EGFR mutation in lung adenocarcinomas. Moreover, BMXΔN increases cell proliferation, neoplastic transformation, and migratory property of human non-small cell lung cancer cells. The function of BMXΔN in lung cancer might be presumably due to enhanced ERK signaling.
Collapse
Affiliation(s)
- Ye Wang
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Jufeng Xia
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Zhaoyuan Fang
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Fei Li
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Duo Li
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Zuoyun Wang
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Yan Feng
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Jian Zhang
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hongbin Ji
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, 200120, China
| | - Hongyan Liu
- CAS Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, 200031, China.,Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, Shandong, China
| |
Collapse
|
7
|
Hsueh SC, Chen KY, Lai JH, Wu CC, Yu YW, Luo Y, Hsieh TH, Chiang YH. Voluntary Physical Exercise Improves Subsequent Motor and Cognitive Impairments in a Rat Model of Parkinson's Disease. Int J Mol Sci 2018; 19:ijms19020508. [PMID: 29419747 PMCID: PMC5855730 DOI: 10.3390/ijms19020508] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/28/2017] [Revised: 01/19/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023] Open
Abstract
Background: Parkinson’s disease (PD) is typically characterized by impairment of motor function. Gait disturbances similar to those observed in patients with PD can be observed in animals after injection of neurotoxin 6-hydroxydopamine (6-OHDA) to induce unilateral nigrostriatal dopamine depletion. Exercise has been shown to be a promising non-pharmacological approach to reduce the risk of neurodegenerative disease. Methods: In this study, we investigated the long-term effects of voluntary running wheel exercise on gait phenotypes, depression, cognitive, rotational behaviors as well as histology in a 6-OHDA-lesioned rat model of PD. Results: We observed that, when compared with the non-exercise controls, five-week voluntary exercise alleviated and postponed the 6-OHDA-induced gait deficits, including a significantly improved walking speed, step/stride length, base of support and print length. In addition, we found that the non-motor functions, such as novel object recognition and forced swim test, were also ameliorated by voluntary exercise. However, the rotational behavior of the exercise group did not show significant differences when compared with the non-exercise group. Conclusions: We first analyzed the detailed spatiotemporal changes of gait pattern to investigate the potential benefits after long-term exercise in the rat model of PD, which could be useful for future objective assessment of locomotor function in PD or other neurological animal models. Furthermore, these results suggest that short-term voluntary exercise is sufficient to alleviate cognition deficits and depressive behavior in 6-OHDA lesioned rats and long-term treatment reduces the progression of motor symptoms and elevates tyrosine hydroxylase (TH), Brain-derived neurotrophic factor (BDNF), bone marrow tyrosine kinase in chromosome X (BMX) protein expression level without affecting dopaminergic (DA) neuron loss in this PD rat model.
Collapse
Affiliation(s)
- Shih-Chang Hsueh
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
| | - Kai-Yun Chen
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jing-Huei Lai
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chung-Che Wu
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Yu-Wen Yu
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu Luo
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Tsung-Hsun Hsieh
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Yung-Hsiao Chiang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| |
Collapse
|
8
|
Wang Q, Zeng F, Sun Y, Qiu Q, Zhang J, Huang W, Huang J, Huang X, Guo L. Etk Interaction with PFKFB4 Modulates Chemoresistance of Small-cell Lung Cancer by Regulating Autophagy. Clin Cancer Res 2017; 24:950-962. [PMID: 29208667 DOI: 10.1158/1078-0432.ccr-17-1475] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/23/2017] [Revised: 10/30/2017] [Accepted: 12/01/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Epithelial and endothelial tyrosine kinase (Etk), also known as bone marrow X kinase (Bmx), was found to be critical in modulating the chemoresistance of small-cell lung cancer (SCLC) in our preliminary study. However, the molecular mechanisms of Etk in SCLC chemoresistance remain poorly understood.Experimental Design: We determined correlation of Etk with autophagy in SCLC. And direct inhibition of autophagy was performed to validate its effect on chemoresistance. Coimmunoprecipitation (co-IP) and GST-pull down experiments were conducted to verify the interaction of Etk and PFKFB4, after a microarray analysis. In vitro and in vivo gain or loss-of-function analyses and evaluation of PFKFB4 expression in SCLC specimens, were done to validate its role in chemoresistance. Ibrutinib was administrated in SCLC cells to verify its synergistic anti-tumor effect with chemotherapy using preclinical models including a PDX model.Results: Downregulation of Etk suppressed autophagy in chemoresistant SCLC cells, and direct inhibition of autophagy sensitized cells to chemotherapy. PFKFB4 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4) was identified as a downstream target of Etk and an Etk-interacting protein, which promoted chemoresistance in SCLC and was associated with poor therapeutic response and prognosis. Furthermore, ibrutinib was found to exhibit a synergistic anti-tumor effect with chemotherapy in targeting Etk.Conclusions: Our results demonstrated for the first time that Etk interacts with PFKFB4 to promote SCLC chemoresistance through regulation of autophagy. Aberrant Etk and PFKFB4 can be predictive factors for the chemotherapy response as well as potential therapeutic targets in SCLC. Clin Cancer Res; 24(4); 950-62. ©2017 AACR.
Collapse
Affiliation(s)
- Qiongyao Wang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Fanrui Zeng
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Yanqin Sun
- Department of Pathology, Guangdong Medical University, Dongguan, P.R. China
| | - Qianqian Qiu
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Weimei Huang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jie Huang
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xiaomin Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, P.R. China.
| |
Collapse
|
9
|
Eldeeb MA, Fahlman RP. Phosphorylation Impacts N-end Rule Degradation of the Proteolytically Activated Form of BMX Kinase. J Biol Chem 2016; 291:22757-22768. [PMID: 27601470 DOI: 10.1074/jbc.m116.737387] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/10/2016] [Revised: 08/24/2016] [Indexed: 11/06/2022] Open
Abstract
Cellular signaling leading to the initiation of apoptosis typically results in the activation of caspases, which in turn leads to the proteolytic generation of protein fragments with new or altered cellular functions. Increasing numbers of reports are demonstrating that the activity of many of these proteolytically activated protein fragments can be attenuated by their selective degradation by the N-end rule pathway. Here we report the first evidence that selective degradation of a caspase product by the N-end rule pathway can be modulated by phosphorylation. We demonstrate that the pro-apoptotic fragment of the bone marrow kinase on chromosome X (BMX) generated by caspase cleavage in the prostate cancer-derived PC3 cell line is metabolically unstable in cells because its N-terminal tryptophan targets it for proteasomal degradation via the N-end rule pathway. In addition, we have demonstrated that phosphorylation of tyrosine 566 relatively inhibits degradation of the C-terminal BMX catalytic fragment, and this phosphorylation is crucial for its pro-apoptotic function. Overall, our results demonstrate that cleaved BMX is a novel N-end rule substrate, and its degradation exhibits a novel interplay between substrate phosphorylation and N-end rule degradation, revealing an increasing complex regulatory network of apoptotic proteolytic signaling cascades.
Collapse
Affiliation(s)
| | - Richard P Fahlman
- From the Departments of Biochemistry and .,Oncology, University of Alberta, Edmonton, Alberta T6J 2H7, Canada
| |
Collapse
|
10
|
Guo W, Liu R, Bhardwaj G, Ma AH, Changou C, Yang JC, Li Y, Feng C, Luo Y, Mazloom A, Sanchez E, Wang Y, Huang W, Patterson R, Evans CP, Lam KS, Kung HJ. CTA095, a novel Etk and Src dual inhibitor, induces apoptosis in prostate cancer cells and overcomes resistance to Src inhibitors. PLoS One 2013; 8:e70910. [PMID: 23967135 PMCID: PMC3744530 DOI: 10.1371/journal.pone.0070910] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/07/2013] [Accepted: 06/25/2013] [Indexed: 01/07/2023] Open
Abstract
Etk is a non-receptor tyrosine kinase, which provides a strong survival signal in human prostate cancer cells. Src, another tyrosine kinase that cross-activates with Etk, has been shown to play an important role in prostate cancer metastasis. Herein, we discovered a new class of Etk inhibitors. Within those inhibitors, CTA095 was identified as a potent Etk and Src dual inhibitor. CTA095 was found to induce autophagy as well as apoptosis in human prostate cancer cells. In addition, CTA095 inhibited HUVEC cell tube formation and “wound healing” of human prostate cancer cells, implying its role in inhibition of angiogenesis and metastasis of human prostate cancer. More interestingly, CTA095 could overcome Src inhibitor resistance in prostate cancer cells. It induces apoptosis in Src inhibitor resistant prostate cancer cells, likely through a mechanism of down regulation of Myc and BCL2. This finding indicates that simultaneously targeting Etk and Src could be a promising approach to overcome drug resistance in prostate cancer.
Collapse
Affiliation(s)
- Wenchang Guo
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
- * E-mail: (RL); (HJK)
| | - Gaurav Bhardwaj
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Chun Changou
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Joy C. Yang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Caihong Feng
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Yan Luo
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Anisha Mazloom
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Eduardo Sanchez
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Yan Wang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Wenzhe Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Randen Patterson
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
- Department of Physiology and Membrane Biology, University of California Davis, Sacramento, California, United States of America
| | - Christopher P. Evans
- Department of Urology, University of California Davis, Sacramento, California, United States of America
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
| | - Hsing-Jien Kung
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, United States of America
- National Health Research Institutes (NHRI), Miaoli County, Taiwan, ROC
- * E-mail: (RL); (HJK)
| |
Collapse
|
11
|
Varshavsky A. Augmented generation of protein fragments during wakefulness as the molecular cause of sleep: a hypothesis. Protein Sci 2012; 21:1634-61. [PMID: 22930402 PMCID: PMC3527701 DOI: 10.1002/pro.2148] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/02/2012] [Accepted: 08/21/2012] [Indexed: 02/05/2023]
Abstract
Despite extensive understanding of sleep regulation, the molecular-level cause and function of sleep are unknown. I suggest that they originate in individual neurons and stem from increased production of protein fragments during wakefulness. These fragments are transient parts of protein complexes in which the fragments were generated. Neuronal Ca²⁺ fluxes are higher during wakefulness than during sleep. Subunits of transmembrane channels and other proteins are cleaved by Ca²⁺-activated calpains and by other nonprocessive proteases, including caspases and secretases. In the proposed concept, termed the fragment generation (FG) hypothesis, sleep is a state during which the production of fragments is decreased (owing to lower Ca²⁺ transients) while fragment-destroying pathways are upregulated. These changes facilitate the elimination of fragments and the remodeling of protein complexes in which the fragments resided. The FG hypothesis posits that a proteolytic cleavage, which produces two fragments, can have both deleterious effects and fitness-increasing functions. This (previously not considered) dichotomy can explain both the conservation of cleavage sites in proteins and the evolutionary persistence of sleep, because sleep would counteract deleterious aspects of protein fragments. The FG hypothesis leads to new explanations of sleep phenomena, including a longer sleep after sleep deprivation. Studies in the 1970s showed that ethanol-induced sleep in mice can be strikingly prolonged by intracerebroventricular injections of either Ca²⁺ alone or Ca²⁺ and its ionophore (Erickson et al., Science 1978;199:1219-1221; Harris, Pharmacol Biochem Behav 1979;10:527-534; Erickson et al., Pharmacol Biochem Behav 1980;12:651-656). These results, which were never interpreted in connection to protein fragments or the function of sleep, may be accounted for by the FG hypothesis about molecular causation of sleep.
Collapse
Affiliation(s)
- Alexander Varshavsky
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA.
| |
Collapse
|
12
|
Chen KY, Wu CC, Chang CF, Chen YH, Chiu WT, Lou YH, Chen YH, Shih HM, Chiang YH. Suppression of Etk/Bmx Protects against Ischemic Brain Injury. Cell Transplant 2012; 21:345-54. [DOI: 10.3727/096368911x582741] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023] Open
Abstract
Etk/Bmx (epithelial and endothelial tyrosine kinase, also known as BMX), a member of the Tec (tyrosine kinase expressed in hepatocellular carcinoma) family of protein-tyrosine kinases, is an important regulator of signal transduction for the activation of cell growth, differentiation, and development. We have previously reported that activation of Etk leads to apoptosis in MDA-MB-468 cells. The purpose of this study was to examine the role of Etk in neuronal injury induced by H2O2 or ischemia. Using Western blot analysis and immunohistochemistry, we found that treatment with H2O2 significantly enhanced phosphorylation of Etk and its downstream signaling molecule Stat1 in primary cortical neurons. Inhibiting Etk activity by LFM-A13 or knocking down Etk expression by a specific shRNA increased the survival of primary cortical neurons. Similarly, at 1 day after a 60-min middle cerebral artery occlusion (MCAo) in adult rats, both phosphorylated Etk and Stat1 were coexpressed with apoptotic markers in neurons in the penumbra. Pretreatment with LFM-A13 or an adenoviral vector encoding the kinase deletion mutant EtkΔk attenuated caspase-3 activity and infarct volume in ischemic brain. All together, our data suggest that Etk is activated after neuronal injury. Suppressing Etk activity protects against neurodegeneration in ischemic brain.
Collapse
Affiliation(s)
- Kai-Yun Chen
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Translational Research Laboratory, Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chung-Che Wu
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Fu Chang
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Ta Chiu
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ya-Hsin Lou
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Hua Chen
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Translational Research Laboratory, Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsiu-Ming Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Translational Research Laboratory, Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
13
|
Varshavsky A. The N-end rule pathway and regulation by proteolysis. Protein Sci 2011; 20:1298-345. [PMID: 21633985 PMCID: PMC3189519 DOI: 10.1002/pro.666] [Citation(s) in RCA: 551] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2011] [Revised: 05/16/2011] [Accepted: 05/18/2011] [Indexed: 01/12/2023]
Abstract
The N-end rule relates the regulation of the in vivo half-life of a protein to the identity of its N-terminal residue. Degradation signals (degrons) that are targeted by the N-end rule pathway include a set called N-degrons. The main determinant of an N-degron is a destabilizing N-terminal residue of a protein. In eukaryotes, the N-end rule pathway is a part of the ubiquitin system and consists of two branches, the Ac/N-end rule and the Arg/N-end rule pathways. The Ac/N-end rule pathway targets proteins containing N(α) -terminally acetylated (Nt-acetylated) residues. The Arg/N-end rule pathway recognizes unacetylated N-terminal residues and involves N-terminal arginylation. Together, these branches target for degradation a majority of cellular proteins. For example, more than 80% of human proteins are cotranslationally Nt-acetylated. Thus most proteins harbor a specific degradation signal, termed (Ac)N-degron, from the moment of their birth. Specific N-end rule pathways are also present in prokaryotes and in mitochondria. Enzymes that produce N-degrons include methionine-aminopeptidases, caspases, calpains, Nt-acetylases, Nt-amidases, arginyl-transferases and leucyl-transferases. Regulated degradation of specific proteins by the N-end rule pathway mediates a legion of physiological functions, including the sensing of heme, oxygen, and nitric oxide; selective elimination of misfolded proteins; the regulation of DNA repair, segregation and condensation; the signaling by G proteins; the regulation of peptide import, fat metabolism, viral and bacterial infections, apoptosis, meiosis, spermatogenesis, neurogenesis, and cardiovascular development; and the functioning of adult organs, including the pancreas and the brain. Discovered 25 years ago, this pathway continues to be a fount of biological insights.
Collapse
Affiliation(s)
- Alexander Varshavsky
- 1Division of Biology, California Institute of Technology, Pasadena, California 91125.
| |
Collapse
|
14
|
Guryanova OA, Wu Q, Cheng L, Lathia JD, Huang Z, Yang J, MacSwords J, Eyler CE, McLendon RE, Heddleston JM, Shou W, Hambardzumyan D, Lee J, Hjelmeland AB, Sloan AE, Bredel M, Stark GR, Rich JN, Bao S. Nonreceptor tyrosine kinase BMX maintains self-renewal and tumorigenic potential of glioblastoma stem cells by activating STAT3. Cancer Cell 2011; 19:498-511. [PMID: 21481791 PMCID: PMC3076106 DOI: 10.1016/j.ccr.2011.03.004] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/11/2010] [Revised: 02/01/2011] [Accepted: 03/01/2011] [Indexed: 01/26/2023]
Abstract
Glioblastomas display cellular hierarchies containing tumor-propagating glioblastoma stem cells (GSCs). STAT3 is a critical signaling node in GSC maintenance but molecular mechanisms underlying STAT3 activation in GSCs are poorly defined. Here we demonstrate that the bone marrow X-linked (BMX) nonreceptor tyrosine kinase activates STAT3 signaling to maintain self-renewal and tumorigenic potential of GSCs. BMX is differentially expressed in GSCs relative to nonstem cancer cells and neural progenitors. BMX knockdown potently inhibited STAT3 activation, expression of GSC transcription factors, and growth of GSC-derived intracranial tumors. Constitutively active STAT3 rescued the effects of BMX downregulation, supporting that BMX signals through STAT3 in GSCs. These data demonstrate that BMX represents a GSC therapeutic target and reinforces the importance of STAT3 signaling in stem-like cancer phenotypes.
Collapse
Affiliation(s)
- Olga A. Guryanova
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Qiulian Wu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lin Cheng
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Experimental Center, The First People’s Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Justin D. Lathia
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Zhi Huang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jinbo Yang
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jennifer MacSwords
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Christine E. Eyler
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Roger E. McLendon
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - John M. Heddleston
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dolores Hambardzumyan
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anita B. Hjelmeland
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew E. Sloan
- Brain Tumor and Neuro-Oncology Center, University Hospitals, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Markus Bredel
- Departments of Radiation Oncology, Genetics, and Cell Biology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35249, USA
| | - George R. Stark
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jeremy N. Rich
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Co-correspondence: 9500 Euclid Avenue, NE30, Cleveland Clinic, Cleveland, OH 44195, USA; Tel: +1 216 636 0790; Fax: +1 216 636 5454;
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Correspondence: 9500 Euclid Avenue, NE30, Cleveland Clinic, Cleveland, OH 44195, USA; Tel: +1 216 636 1009; Fax: +1 216 636 5454;
| |
Collapse
|
15
|
Abstract
Tyrosine kinases play significant roles in tumor progression and therapy resistance. Inhibitors of tyrosine kinases are on the forefront of targeted therapy. For prostate cancer, tyrosine kinases play an additional role in the development of castration-resistant disease state, the most troubling aspect of prostate cancinogenesis which presently defies any effective treatment. Among the 30 or so tyrosine kinases expressed in a typical prostate cancer cell, nearly one third of them have been implicated in prostate carcinogenesis. Interestingly, most of them channel signals through a trio of non-receptor tyrosine kinases, Src/Etk/FAK, referred here as Src tyrosine kinase complex. This complex has been shown to play a significant role in the aberrant activation of androgen receptor (AR) mediated by growth factors (e.g., epidermal growth factor (EGF)), cytokines (interleukin (IL)-6), chemokines (IL-8), and neurokines (gastrin-releasing peptide). These factors are induced and released from the prostate cancer to the stromal cells upon androgen withdrawal. The Src kinase complex has the ability to phosphorylate androgen receptor, resulting in the nuclear translocation and stabilization of un-liganded androgen receptor. Indeed, tyrosine kinase inhibitors targeting Src can inhibit androgen-independent growth of prostate cancer cells in vitro and in preclinical xenograft model. While effective in inducing growth arrest and inhibiting metastasis of castration-resistant tumors, Src inhibitors rarely induce a significant level of apoptosis. This is also reflected by the general ineffectiveness of tyrosine kinase inhibitors as monotherapy in clinical trials. One of the underlying causes of apoptosis resistance is "autophagy," which is induced by tyrosine kinase inhibitors and by androgen withdrawal. Autophagy is a self-digesting process to regenerate energy by removal of long-lived proteins and retired organelles to provide a survival mechanism to cells encountering stresses. Excessive autophagy, sometimes, could lead to type II programmed cell death. We demonstrated that autophagy blockade sensitizes prostate cancer cells toward Src tyrosine kinase inhibitor. Thus, a combination therapy based on Src tyrosine kinase inhibitor and autophagy modulator deserves further attention as a potential treatment for relapsed prostate cancer.
Collapse
Affiliation(s)
- Hsing-Jien Kung
- UC Davis Cancer Center, UCDMC, Res III, Rm. 2400, 4645 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
16
|
Abstract
BACKGROUND Caspases belong to a class of cysteine proteases which function as critical effectors in cellular processes such as apoptosis and inflammation by cleaving substrates immediately after unique tetrapeptide sites. With hundreds of reported substrates and many more expected to be discovered, the elucidation of the caspase degradome will be an important milestone in the study of these proteases in human health and disease. Several computational methods for predicting caspase cleavage sites have been developed recently for identifying potential substrates. However, as most of these methods are based primarily on the detection of the tetrapeptide cleavage sites - a factor necessary but not sufficient for predicting in vivo substrate cleavage - prediction outcomes will inevitably include many false positives. RESULTS In this paper, we show that structural factors such as the presence of disorder and solvent exposure in the vicinity of the cleavage site are important and can be used to enhance results from cleavage site prediction. We constructed a two-step model incorporating cleavage site prediction and these factors to predict caspase substrates. Sequences are first predicted for cleavage sites using CASVM or GraBCas. Predicted cleavage sites are then scored, ranked and filtered against a cut-off based on their propensities for locating in disordered and solvent exposed regions. Using an independent dataset of caspase substrates, the model was shown to achieve greater positive predictive values compared to CASVM or GraBCas alone, and was able to reduce the false positives pool by up to 13% and 53% respectively while retaining all true positives. We applied our prediction model on the family of receptor tyrosine kinases (RTKs) and highlighted several members as potential caspase targets. The results suggest that RTKs may be generally regulated by caspase cleavage and in some cases, promote the induction of apoptotic cell death - a function distinct from their role as transducers of survival and growth signals. CONCLUSION As a step towards the prediction of in vivo caspase substrates, we have developed an accurate method incorporating cleavage site prediction and structural factors. The multi-factor model augments existing methods and complements experimental efforts to define the caspase degradome on the systems-wide basis.
Collapse
Affiliation(s)
- Lawrence J K Wee
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | | | | |
Collapse
|
17
|
Mitchell-Jordan SA, Holopainen T, Ren S, Wang S, Warburton S, Zhang MJ, Alitalo K, Wang Y, Vondriska TM. Loss of Bmx nonreceptor tyrosine kinase prevents pressure overload-induced cardiac hypertrophy. Circ Res 2008; 103:1359-62. [PMID: 18988895 DOI: 10.1161/circresaha.108.186577] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Bmx nonreceptor tyrosine kinase has an established role in endothelial and lymphocyte signaling; however, its role in the heart is unknown. To determine whether Bmx participates in cardiac growth, we subjected mice deficient in the molecule (Bmx knockout mice) to transverse aortic constriction (TAC). In comparison with wild-type mice, which progressively developed massive hypertrophy following TAC, Bmx knockout mice were resistant to TAC-induced cardiac growth at the organ and cell level. Loss of Bmx preserved cardiac ejection fraction and decreased mortality following TAC. These findings are the first to demonstrate a necessary role for the Tec family of tyrosine kinases in the heart and reveal a novel regulator (Bmx) of pressure overload-induced hypertrophic growth.
Collapse
Affiliation(s)
- Scherise A Mitchell-Jordan
- Department of Anesthesiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Choi MR, Groot M, Drexler HCA. Functional implications of caspase-mediated RhoGDI2 processing during apoptosis of HL60 and K562 leukemia cells. Apoptosis 2007; 12:2025-35. [PMID: 17726646 DOI: 10.1007/s10495-007-0121-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
RhoGDI2, a cytosolic regulator of Rho GTPase, is cleaved during apoptosis in a caspase-3 dependent fashion. By using 2D-gel electrophoresis, mass spectrometry and Western blotting we investigate in this paper the functional consequences of RhoGDI2 processing. We can show that loss of the N-terminal 19 amino acids results in a shift of the isoelectric point of the truncated RhoGDI2 (NDelta19) to a more basic value due to the removal of 9 acidic amino acids from the N-terminus, which may be responsible for enhanced retention of the N-terminally truncated protein within the nuclear compartment. Fusion of the p53 nuclear export signaling sequence MFRELNEALELK to NDelta19 (NDelta19NES) abolished its apoptosis promoting properties, while overexpression of NDelta19 significantly increased the susceptibility to apoptosis induction by the proteasome inhibitor PSI and by staurosporine. These results suggest that cleavage of RhoGDI2 by caspase-3 is not a functionally irrelevant bystander effect of caspase activation during apoptosis, but rather expedites progression of the apoptotic process.
Collapse
MESH Headings
- Amino Acid Sequence
- Apoptosis/genetics
- Apoptosis/physiology
- Caspases/physiology
- Electrophoresis, Gel, Two-Dimensional
- Guanine Nucleotide Dissociation Inhibitors/genetics
- Guanine Nucleotide Dissociation Inhibitors/metabolism
- HL-60 Cells
- Humans
- K562 Cells
- Leukemia, Erythroblastic, Acute/metabolism
- Leukemia, Erythroblastic, Acute/pathology
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Molecular Sequence Data
- RNA Processing, Post-Transcriptional
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Tumor Cells, Cultured
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- U937 Cells
- rho Guanine Nucleotide Dissociation Inhibitor beta
- rho-Specific Guanine Nucleotide Dissociation Inhibitors
Collapse
Affiliation(s)
- Mi-Ran Choi
- Max-Planck-Institute for Heart and Lung Research, Parkstr.1, Bad Nauheim 61231, Germany
| | | | | |
Collapse
|
19
|
Abstract
BACKGROUND Carcinoma of the prostate (CaP) is the most commonly diagnosed cancer in men in the United States. Signal transduction molecules such as tyrosine kinases play important roles in CaP. Src, a nonreceptor tyrosine kinase (NRTK) and the first proto-oncogene discovered is shown to participate in processes such as cell proliferation and migration in CaP. Underscoring NRTK's and, specifically, Src's importance in cancer is the recent approval by the US Food and Drug Administration of dasatinib, the first commercial Src inhibitor for clinical use in chronic myelogenous leukemia (CML). In this review we will focus on NRTKs and their roles in the biology of CaP. MATERIALS AND METHODS Publicly available literature from PubMed regarding the topic of members of NRTKs in CaP was searched and reviewed. RESULTS Src, FAK, JaK1/2, and ETK are involved in processes indispensable to the biology of CaP: cell growth, migration, invasion, angiogenesis, and apoptosis. CONCLUSIONS Src emerges as a common signaling and regulatory molecule in multiple biological processes in CaP. Src's relative importance in particular stages of CaP, however, required further definition. Continued investigation of NRTKs will increase our understanding of their biological function and potential role as new therapeutic targets.
Collapse
Affiliation(s)
- Yu-Ming Chang
- Department of Urology, University of California at Davis, Sacramento, CA 95817, USA
| | | | | |
Collapse
|
20
|
Abstract
The receptor tyrosine kinase ErbB-2 plays an important role in cell proliferation and differentiation as well as oncogenesis. We have found that ErbB-2 kinase domain fragmentation is important for the induction of apoptosis. Exogenous expression of peptides derived from the ErbB-2 kinase domain induces cells death with the hallmarks of apoptosis. In contrast, transfection of the ErbB-2 carboxy-terminal domain did not induce apoptosis. We have identified a 37-residue segment from the ErbB-2 kinase N-terminal lobe that can strongly induce apoptosis in transfected cells. Cell death was not blocked by the pan-caspase inhibitor z-VAD-FMK. Similar fragments derived from several other receptor tyrosine kinases also induce cell death. These data imply that proteolytic fragmentation of tyrosine kinases liberates apoptotic fragments that can accelerate cell death.
Collapse
Affiliation(s)
- Oleg Tikhomirov
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| | | | | |
Collapse
|
21
|
Wu JC, Chen TY, Yu CTR, Tsai SJ, Hsu JM, Tang MJ, Chou CK, Lin WJ, Yuan CJ, Huang CYF. Identification of V23RalA-Ser194 as a critical mediator for Aurora-A-induced cellular motility and transformation by small pool expression screening. J Biol Chem 2005; 280:9013-22. [PMID: 15637052 DOI: 10.1074/jbc.m411068200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/04/2023] Open
Abstract
Human Aurora kinases have three gene family members: Aurora-A, Aurora-B, and Aurora-C. It is not yet established what the specificity of these kinases are and what signals relayed by their reactions. Therefore, we employed small pool expression screening to search for downstream substrates of Aurora-A. Interestingly, all of the identified Aurora-A substrates were resistant to serve as substrates for Aurora-B or Aurora-C, suggesting that these Aurora family members may have distinct substrate specificity for propagation of diverse signaling pathways, even though they share a conserved catalytic kinase domain. Of the candidate substrates, Aurora-A could increase the functional activity of RalA. Mutational analysis revealed that RalA-Ser194 was the phosphorylation site for Aurora-A. Ectopic expression of V23RalA-WT could enhance collagen I-induced cell migration and anchorage-independent growth in Madin-Darby canine kidney (MDCK) Aurora-A stable cell lines. In contrast, overexpression of V23RalA-S194A in MDCK Aurora-A-stable cell lines abolished the intrinsic migration and transformation abilities of Aurora-A. To our knowledge, this is the first systematic search for the downstream substrates of Aurora-A kinase. Moreover, these results support the notion that Aurora-A may act in concert with V23RalA through protein phosphorylation on Ser194 to promote collagen I-induced cell motility and anchorage-independent growth in MDCK epithelial cells.
Collapse
Affiliation(s)
- Jiunn-Chyi Wu
- Division of Molecular and Genomic Medicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County 350, Taipei, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Stoletov KV, Terman BI. Bmx is a downstream Rap1 effector in VEGF-induced endothelial cell activation. Biochem Biophys Res Commun 2004; 320:70-5. [PMID: 15207703 DOI: 10.1016/j.bbrc.2004.05.134] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/17/2004] [Indexed: 11/25/2022]
Abstract
We had previously shown that Rap1 mediates certain of the signaling pathways involved in VEGF-induced endothelial cell migration, although the downstream Rap1 effectors are not known. Towards the goal of identifying those effectors, we utilized a commercially available antibody array filter to identify proteins that either directly interact with Rap1 or interact indirectly through a multi-protein complex. The protocol identified 10 possible Rap1-interacting proteins, including the Bmx non-receptor tyrosine kinase. The conclusion that VEGF treatment leads to a Rap1/Bmx complex was confirmed by an experiment in which cell lysates from VEGF and control cells were immunoprecipitated with Bmx antibodies and Western blotting was done using anti-Rap1 antibodies. VEGF treatment led to the recruitment of Bmx to the CAS scaffolding protein, and inhibition of the Bmx kinase blocked VEGF-induced cell migration. Formation of a Rap1/Bmx complex was not observed in cells transfected with an expression vector for a dominant-negative Rap1, indicating that Bmx is a downstream Rap1 effector in VEGF-induced endothelial cell activation.
Collapse
Affiliation(s)
- Konstantin V Stoletov
- Cardiology Division, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
23
|
Chen KY, Huang LM, Kung HJ, Ann DK, Shih HM. The role of tyrosine kinase Etk/Bmx in EGF-induced apoptosis of MDA-MB-468 breast cancer cells. Oncogene 2004; 23:1854-62. [PMID: 14676838 DOI: 10.1038/sj.onc.1207308] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/09/2022]
Abstract
Etk/Bmx, a member of the Tec family of tyrosine kinases, mediates various signaling pathways and confers several cellular functions. In the present study, we have explored the functional role of Etk in mediating EGF-induced apoptosis, using MDA-MB-468 cell line as a model. We first demonstrated that EGF treatment induces Etk tyrosine phosphorylation in both HeLa and MDA-MB-468 cells. Overexpression of Etk by recombinant adenovirus in MDA-MB-468 cells potentiates the extent of EGF-induced cell apoptosis. The observed Etk-enhanced MDA-MB-468 cell apoptosis is associated with the Stat1 activation, as demonstrated by electrophoresis mobility shift assays and reporter gene assays. By contrast, a kinase domain deletion mutant EtkDeltaK, functioning as a dominant-negative mutant, ameliorates EGF-induced Stat1 activation and apoptosis in MDA-MB-468 cells. To explore whether the activated Etk alone is sufficient for inducing apoptosis, a conditionally activated Etk (DeltaEtk-ER), a chimeric fusion protein of PH domain-truncated Etk and ligand-binding domain of estrogen receptor, was introduced into MDA-MB-468 cells. Upon beta-estradiol ligand activation, the DeltaEtk-ER could stimulate Stat1 activity and confer cell apoptosis independent of EGF treatment. Taken together, our findings indicate that Etk is a downstream signaling molecule of EGF receptor and suggest that Etk activation is essential for transducing the EGF-induced apoptotic signaling.
Collapse
Affiliation(s)
- Kai-Yun Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
24
|
Hur YG, Yun Y, Won J. Rosmarinic acid induces p56lck-dependent apoptosis in Jurkat and peripheral T cells via mitochondrial pathway independent from Fas/Fas ligand interaction. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2004; 172:79-87. [PMID: 14688312 DOI: 10.4049/jimmunol.172.1.79] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2023]
Abstract
Apoptosis is one way of controlling immune responses, and a variety of immunosuppressive drugs suppress harmful immune responses by inducing apoptosis of lymphocytes. In this study we observed that rosmarinic acid, a secondary metabolite of herbal plants, induced apoptosis in an p56(lck) (Lck)-dependent manner; Lck(+) Jurkat T cells undergo apoptosis in response to rosmarinic acid (RosA) treatment, whereas Lck(-) Jurkat subclone J.CaM1.6 cells do not. J.CaM1.6 cells with various Lck mutants indicated that Lck SH2 domain, but not Lck kinase activity, was required for RosA-induced apoptosis. RosA induced apoptosis in the absence of a TCR stimulus, and this was not prevented by interruption of the Fas/Fas ligand interaction. Instead, RosA-mediated apoptosis involved a mitochondrial pathway as indicated by cytochrome c release and the complete blockage of apoptosis by an inhibitor of mitochondrial membrane depolarization. Both caspase-3 and -8 were indispensable in RosA-induced apoptosis and work downstream of mitochondria and caspase-9 in the order of caspase-9/caspase-3/caspase-8. In freshly isolated human PBMC, RosA specifically induced apoptosis of Lck(+) subsets such as T and NK cells, but not Lck-deficient cells, including B cells and monocytes. Moreover, RosA's ability to kill T and NK cells was restricted to actively proliferating cells, but not to resting cells. In conclusion, Lck-dependent apoptotic activity may make RosA an attractive therapeutic tool for the treatment of diseases in which T cell apoptosis is beneficial.
Collapse
Affiliation(s)
- Yun-Gyoung Hur
- Mogam Biotechnology Research Institute, 304 Bojungri, Goosungeup, Yongin City, Gyunggido 449-913, Korea
| | | | | |
Collapse
|
25
|
Barilà D, Rufini A, Condò I, Ventura N, Dorey K, Superti-Furga G, Testi R. Caspase-dependent cleavage of c-Abl contributes to apoptosis. Mol Cell Biol 2003; 23:2790-9. [PMID: 12665579 PMCID: PMC152541 DOI: 10.1128/mcb.23.8.2790-2799.2003] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
The nonreceptor tyrosine kinase c-Abl may contribute to the regulation of apoptosis. c-Abl activity is induced in the nucleus upon DNA damage, and its activation is required for execution of the apoptotic program. Recently, activation of nuclear c-Abl during death receptor-induced apoptosis has been reported; however, the mechanism remains largely obscure. Here we show that c-Abl is cleaved by caspases during tumor necrosis factor- and Fas receptor-induced apoptosis. Cleavage at the very C-terminal region of c-Abl occurs mainly in the cytoplasmic compartment and generates a 120-kDa fragment that lacks the nuclear export signal and the actin-binding region but retains the intact kinase domain, the three nuclear localization signals, and the DNA-binding domain. Upon caspase cleavage, the 120-kDa fragment accumulates in the nucleus. Transient-transfection experiments show that cleavage of c-Abl may affect the efficiency of Fas-induced cell death. These data reveal a novel mechanism by which caspases can recruit c-Abl to the nuclear compartment and to the mammalian apoptotic program.
Collapse
Affiliation(s)
- Daniela Barilà
- Dulbecco Telethon Institute and Laboratory of Immunology and Signal Transduction, Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Apoptotic cell death is executed by the caspase-mediated cleavage of various vital proteins. Elucidating the consequences of this endoproteolytic cleavage is crucial for our understanding of cell death and other biological processes. Many caspase substrates are just cleaved as bystanders, because they happen to contain a caspase cleavage site in their sequence. Several targets, however, have a discrete function in propagation of the cell death process. Many structural and regulatory proteins are inactivated by caspases, while other substrates can be activated. In most cases, the consequences of this gain-of-function are poorly understood. Caspase substrates can regulate the key morphological changes in apoptosis. Several caspase substrates also act as transducers and amplifiers that determine the apoptotic threshold and cell fate. This review summarizes the known caspase substrates comprising a bewildering list of more than 280 different proteins. We highlight some recent aspects inferred by the cleavage of certain proteins in apoptosis. We also discuss emerging themes of caspase cleavage in other forms of cell death and, in particular, in apparently unrelated processes, such as cell cycle regulation and cellular differentiation.
Collapse
Affiliation(s)
- U Fischer
- Institute of Molecular Medicine, University of Düsseldorf, Germany
| | - R U Jänicke
- Institute of Molecular Medicine, University of Düsseldorf, Germany
| | | |
Collapse
|
27
|
Chau CH, Chen KY, Deng HT, Kim KJ, Hosoya KI, Terasaki T, Shih HM, Ann DK. Coordinating Etk/Bmx activation and VEGF upregulation to promote cell survival and proliferation. Oncogene 2002; 21:8817-29. [PMID: 12483534 DOI: 10.1038/sj.onc.1206032] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/21/2002] [Revised: 08/30/2002] [Accepted: 09/05/2002] [Indexed: 11/09/2022]
Abstract
Etk/Bmx, a member of the Tec family of non-receptor tyrosine kinase, is characterized by an N-terminal PH domain and has recently been shown to be involved in the regulation of various cellular processes, including proliferation, differentiation, motility and apoptosis. Since VEGF and the activation of its signaling pathway have been implicated in modulating a variety of biological responses, we characterized the role of Etk-dependent signaling pathways involved in the upregulation of VEGF expression, and explored the functional implications of this enhancement in sustaining cell proliferation and survival. Using Northern and Western analyses, transient transfections, and pharmacological agents, we demonstrate that Etk activation alone is sufficient to transcriptionally induce VEGF expression, independent of the previously identified hypoxia response element (HRE), in both Pa-4 epithelial and TR-BBB endothelial cells under normoxia. In addition, Etk utilizes both MEK/ERK and PI3-K/Pak1 signaling pathways in concert to activate VEGF transcription. Functionally, Etk activation elicits a profound stimulatory effect on TR-BBB cell proliferation and formation of capillary-like networks in Matrigel containing reduced levels of growth factors. Finally, antisense oligonucleotides against either endogenous VEGF or Etk abrogate the proliferation of Etk-activated TR-BBB cells, and exogenous VEGF treatment stimulates endogenous Etk tyrosine phosphorylation in HUVECs. Taken together, these results indicate that VEGF is both an Etk downstream target gene and an Etk upstream activator, constituting a reciprocal Etk-VEGF autoregulatory loop. These findings, to our knowledge, are the first delineation of a network of positive feedforward signaling pathways that converge on the Etk-VEGF axis, causally associating Etk-mediation of VEGF induction with enhanced cellular processes in both epithelial and endothelial cells.
Collapse
Affiliation(s)
- Cindy H Chau
- Department of Molecular Pharmacology and Toxicology, University of Southern California, Los Angeles, California, CA 90033, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Huang CYF, Wu YM, Hsu CY, Lee WS, Lai MD, Lu TJ, Huang CL, Leu TH, Shih HM, Fang HI, Robinson DR, Kung HJ, Yuan CJ. Caspase activation of mammalian sterile 20-like kinase 3 (Mst3). Nuclear translocation and induction of apoptosis. J Biol Chem 2002; 277:34367-74. [PMID: 12107159 DOI: 10.1074/jbc.m202468200] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian Sterile 20-like kinase 3 (Mst3), the physiological functions of which are unknown, is a member of the germinal center kinase-III family. It contains a conserved kinase domain at its NH(2) terminus, whereas there is a regulatory domain at its COOH terminus. In this study we demonstrate that endogenous Mst3 is specifically cleaved when Jurkat cells were treated with anti-Fas antibody or staurosporine and that this cleavage is inhibited by the caspase inhibitor, Ac-DEVD-CHO. Using apoptotic Jurkat cell extracts and recombinant caspases, we mapped the caspase cleavage site, AETD(313), which is at the junction of the NH(2)-terminal kinase domain and the COOH-terminal regulatory domain. Caspase-mediated cleavage of Mst3 activates its intrinsic kinase activity, suggesting that the COOH-terminal domain of Mst3 negatively regulates the kinase domain. Furthermore, proteolytic removal of the Mst3 COOH-terminal domain by caspases promotes nuclear translocation. Ectopic expression of either wild-type or COOH-terminal truncated Mst3 in cells results in DNA fragmentation and morphological changes characteristic of apoptosis. By contrast, no such changes were exhibited for catalytically inactive Mst3, implicating the involvement of Mst3 kinase activity for mediation of these effects. Collectively, these results support the notion that caspase-mediated proteolytic activation of Mst3 contributes to apoptosis.
Collapse
Affiliation(s)
- Chi-Ying F Huang
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po Ai Street, Hsinchu 300, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|