1
|
Tsai HJ, Chang YF, Hsieh YJ, Wang JD, Wu CC, Ho MY, Cheng JC, Chen DP, Liao HR, Tseng CP. Human Disabled-2 regulates thromboxane A 2 signaling for efficient hemostasis in thrombocytopenia. Nat Commun 2024; 15:9816. [PMID: 39537612 PMCID: PMC11561248 DOI: 10.1038/s41467-024-54093-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding platelet protein functions facilitates better assessment of platelet disorders. Megakaryocyte lineage-restricted human Disabled-2 knock-in (hDAB2-KI) mice are generated to delineate the functions of hDab2, a regulator of platelet function, in the control of bleeding associated with thrombocytopenia. Here we show that hDab2-KI mice with thrombocytopenia display decreased bleeding time when compared to the control mice. hDab2 augments thromboxane A2 (TxA2) mimetic U46619- but not other agonists-stimulated granule secretion, integrin activation, and aggregation at a lower platelet concentration in vitro. Binding of hDab2 to phosphatidic acid (PA) facilitates formation of the PA-hDab2-AKT complex leading to an increase in U46619-stimulated AKT-Ser473 phosphorylation and the first wave of ADP/ATP release. Consistent with these findings, hDab2 expression in platelets from patients with immune thrombocytopenic purpura is positively correlated with U46619-stimulated ATP release, which in turn inversely correlated with their bleeding tendency. hDab2 appears crucial in regulating bleeding severity associated with thrombocytopenia by a functional interplay with ADP/ATP release underlying TxA2 signaling.
Collapse
Affiliation(s)
- Hui-Ju Tsai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ya-Fang Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ya-Ju Hsieh
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, 407, Taiwan, Republic of China
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, 402, Taiwan, Republic of China
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Meng-Ying Ho
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, North District, Taichung, 404, Taiwan, Republic of China
| | - Ding-Ping Chen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
| | - Hsiang-Rui Liao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China.
| |
Collapse
|
2
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Balasooriya ER, Madhusanka D, López-Palacios TP, Eastmond RJ, Jayatunge D, Owen JJ, Gashler JS, Egbert CM, Bulathsinghalage C, Liu L, Piccolo SR, Andersen JL. Integrating Clinical Cancer and PTM Proteomics Data Identifies a Mechanism of ACK1 Kinase Activation. Mol Cancer Res 2024; 22:137-151. [PMID: 37847650 PMCID: PMC10831333 DOI: 10.1158/1541-7786.mcr-23-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/17/2023] [Accepted: 10/13/2023] [Indexed: 10/19/2023]
Abstract
Beyond the most common oncogenes activated by mutation (mut-drivers), there likely exists a variety of low-frequency mut-drivers, each of which is a possible frontier for targeted therapy. To identify new and understudied mut-drivers, we developed a machine learning (ML) model that integrates curated clinical cancer data and posttranslational modification (PTM) proteomics databases. We applied the approach to 62,746 patient cancers spanning 84 cancer types and predicted 3,964 oncogenic mutations across 1,148 genes, many of which disrupt PTMs of known and unknown function. The list of putative mut-drivers includes established drivers and others with poorly understood roles in cancer. This ML model is available as a web application. As a case study, we focused the approach on nonreceptor tyrosine kinases (NRTK) and found a recurrent mutation in activated CDC42 kinase-1 (ACK1) that disrupts the Mig6 homology region (MHR) and ubiquitin-association (UBA) domains on the ACK1 C-terminus. By studying these domains in cultured cells, we found that disruption of the MHR domain helps activate the kinase while disruption of the UBA increases kinase stability by blocking its lysosomal degradation. This ACK1 mutation is analogous to lymphoma-associated mutations in its sister kinase, TNK1, which also disrupt a C-terminal inhibitory motif and UBA domain. This study establishes a mut-driver discovery tool for the research community and identifies a mechanism of ACK1 hyperactivation shared among ACK family kinases. IMPLICATIONS This research identifies a potentially targetable activating mutation in ACK1 and other possible oncogenic mutations, including PTM-disrupting mutations, for further study.
Collapse
Affiliation(s)
- Eranga R. Balasooriya
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Dept. of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Deshan Madhusanka
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Tania P. López-Palacios
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Riley J. Eastmond
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Dasun Jayatunge
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jake J. Owen
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Jack S. Gashler
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Christina M. Egbert
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | | | - Lu Liu
- Department of Computer Science, North Dakota State University, Fargo, North Dakota
| | | | - Joshua L. Andersen
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
4
|
Nawarathnage S, Tseng YJ, Soleimani S, Smith T, Pedroza Romo MJ, Abiodun WO, Egbert CM, Madhusanka D, Bunn D, Woods B, Tsubaki E, Stewart C, Brown S, Doukov T, Andersen JL, Moody JD. Fusion crystallization reveals the behavior of both the 1TEL crystallization chaperone and the TNK1 UBA domain. Structure 2023; 31:1589-1603.e6. [PMID: 37776857 PMCID: PMC10843481 DOI: 10.1016/j.str.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 09/04/2023] [Indexed: 10/02/2023]
Abstract
Human thirty-eight-negative kinase-1 (TNK1) is implicated in cancer progression. The TNK1 ubiquitin-associated (UBA) domain binds polyubiquitin and plays a regulatory role in TNK1 activity and stability. No experimentally determined molecular structure of this unusual UBA domain is available. We fused the UBA domain to the 1TEL variant of the translocation ETS leukemia protein sterile alpha motif (TELSAM) crystallization chaperone and obtained crystals diffracting as far as 1.53 Å. GG and GSGG linkers allowed the UBA to reproducibly find a productive binding mode against its host 1TEL polymer and crystallize at protein concentrations as low as 0.2 mg/mL. Our studies support a mechanism of 1TEL fusion crystallization and show that 1TEL fusion crystals require fewer crystal contacts than traditional protein crystals. Modeling and experimental validation suggest the UBA domain may be selective for both the length and linkages of polyubiquitin chains.
Collapse
Affiliation(s)
| | - Yi Jie Tseng
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Sara Soleimani
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Tobin Smith
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Maria J Pedroza Romo
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Wisdom O Abiodun
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Christina M Egbert
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA; Fritz B. Burns Cancer Research Laboratory, Brigham Young University, Provo, UT, USA
| | - Deshan Madhusanka
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA; Fritz B. Burns Cancer Research Laboratory, Brigham Young University, Provo, UT, USA
| | - Derick Bunn
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Bridger Woods
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Evan Tsubaki
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Cameron Stewart
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Seth Brown
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Tzanko Doukov
- Macromolecular Crystallography Group, Structural Molecular Biology Resource, Stanford Synchrotron Radiation Lightsource, Menlo Park, CA, USA
| | - Joshua L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA; Fritz B. Burns Cancer Research Laboratory, Brigham Young University, Provo, UT, USA.
| | - James D Moody
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
5
|
Fang Y, Bai Z, Cao J, Zhang G, Li X, Li S, Yan Y, Gao P, Kong X, Zhang Z. Low-intensity ultrasound combined with arsenic trioxide induced apoptosis of glioma via EGFR/AKT/mTOR. Life Sci 2023; 332:122103. [PMID: 37730111 DOI: 10.1016/j.lfs.2023.122103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
AIMS This study aimed to explore whether low-intensity ultrasound (LIUS) combined with low-concentration arsenic trioxide (ATO) could inhibit the proliferation of glioma and, if so, to clarify the potential mechanism. MAIN METHODS The effects of ATO and LIUS alone or in combination on glioma were examined by CCK8, EdU, and flow cytometry assays. Western blot analysis was used to detect changes in expression of apoptosis-related proteins and their effects on the EGFR/AKT/mTOR pathway. The effects of ATO and LIUS were verified in vivo in orthotopic xenograft models, and tumor size, arsenic content in brain tissue, survival, and immunohistochemical changes were observed. KEY FINDINGS LIUS enhanced the inhibitory effect of ATO on the proliferation of glioma, and EGF reversed the proliferation inhibition and protein changes induced by ATO and LIUS. The anti-glioma effect of ATO combined with LIUS was related to downstream AKT/mTOR pathway changes caused by inhibition of EGFR activation, which enhanced apoptosis of U87MG and U373 cells. In vivo experiments showed significant increases in arsenic content in brain tissue, as well as decreased tumor sizes and longer survival times in the combined treatment group compared with other groups. The trends of immunohistochemical protein changes were consistent with the in vitro results. SIGNIFICANCE This study showed that LIUS enables ATO to exert anti-glioma effects at a safe dose by inhibiting the activation of EGFR and the downstream AKT/mTOR pathway to regulate apoptosis. LIUS in combination with ATO is a promising novel method for treating glioma and could improve patient prognosis.
Collapse
Affiliation(s)
- Yi Fang
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Zhiqun Bai
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Jibin Cao
- Department of Radiology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Gaosen Zhang
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xiang Li
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Shufeng Li
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Yudie Yan
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Peirong Gao
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiangkai Kong
- Department of Ultrasound, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Zhen Zhang
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Hayashi SY, Craddock BP, Miller WT. Phosphorylation of Ack1 by the Receptor Tyrosine Kinase Mer. KINASES AND PHOSPHATASES 2023; 1:167-180. [PMID: 37662484 PMCID: PMC10473914 DOI: 10.3390/kinasesphosphatases1030011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Ack1 is a nonreceptor tyrosine kinase that is associated with cellular proliferation and survival. The receptor tyrosine kinase Mer, a member of the TAM family of receptors, has previously been reported to be an upstream activator of Ack1 kinase. The mechanism linking the two kinases, however, has not been investigated. We confirmed that Ack1 and Mer interact by co-immunoprecipitation experiments and found that Mer expression led to increased Ack1 activity. The effect on Ack1 was dependent on the kinase activity of Mer, whereas mutation of the Mer C-terminal tyrosines Y867 and Y924 did not significantly decrease the ability of Mer to activate Ack1. Ack1 possesses a Mig6 Homology Region (MHR) that contains adjacent regulatory tyrosines (Y859 and Y860). Using synthetic peptides, we showed that Mer preferentially binds and phosphorylates the MHR sequence containing phosphorylated pY860, as compared to the pY859 sequence. This suggested the possibility of sequential phosphorylation within the MHR of Ack1, as has been observed previously for other kinases. In cells co-expressing Mer and Ack1 MHR mutants, the Y859F mutant had higher activity than the Y860F mutant, consistent with this model. The interaction between Mer and Ack1 could play a role in immune cell signaling in normal physiology and could also contribute to the hyperactivation of Ack1 in prostate cancer and other tumors.
Collapse
Affiliation(s)
- Samantha Y. Hayashi
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Barbara P. Craddock
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Veterans Affairs Medical Center, Northport, NY 11768, USA
| |
Collapse
|
7
|
López-Palacios TP, Andersen JL. Kinase regulation by liquid-liquid phase separation. Trends Cell Biol 2023; 33:649-666. [PMID: 36528418 PMCID: PMC10267292 DOI: 10.1016/j.tcb.2022.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022]
Abstract
Liquid-liquid phase separation (LLPS) is emerging as a mechanism of spatiotemporal regulation that could answer long-standing questions about how order is achieved in biochemical signaling. In this review we discuss how LLPS orchestrates kinase signaling, either by creating condensate structures that are sensed by kinases or by direct LLPS of kinases, cofactors, and substrates - thereby acting as a mechanism to compartmentalize kinase-substrate relationships, and in some cases also sequestering the kinase away from inhibitory factors. We also examine the possibility that selective pressure promotes genomic rearrangements that fuse pro-growth kinases to LLPS-prone protein sequences, which in turn drives aberrant kinase activation through LLPS.
Collapse
Affiliation(s)
- Tania P López-Palacios
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Joshua L Andersen
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
8
|
Nawarathnage S, Tseng YJ, Soleimani S, Smith T, Romo MJP, Abiodun WO, Egbert CM, Madhusanka D, Bunn D, Woods B, Tsubaki E, Stewart C, Brown S, Doukov T, Andersen JL, Moody JD. Fusion crystallization reveals the behavior of both the 1TEL crystallization chaperone and the TNK1 UBA domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.544429. [PMID: 37398013 PMCID: PMC10312729 DOI: 10.1101/2023.06.14.544429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Human thirty-eight-negative kinase-1 (TNK1) is implicated in cancer progression. The TNK1-UBA domain binds polyubiquitin and plays a regulatory role in TNK1 activity and stability. Sequence analysis suggests an unusual architecture for the TNK1 UBA domain, but an experimentally-validated molecular structure is undetermined. To gain insight into TNK1 regulation, we fused the UBA domain to the 1TEL crystallization chaperone and obtained crystals diffracting as far as 1.53 Å. A 1TEL search model enabled solution of the X-ray phases. GG and GSGG linkers allowed the UBA to reproducibly find a productive binding mode against its host 1TEL polymer and to crystallize at protein concentrations as low as 0.1 mg/mL. Our studies support a mechanism of TELSAM fusion crystallization and show that TELSAM fusion crystals require fewer crystal contacts than traditional protein crystals. Modeling and experimental validation suggest the UBA domain may be selective for both the length and linkages of polyubiquitin chains.
Collapse
Affiliation(s)
- Supeshala Nawarathnage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
- These authors contributed equally to this work
| | - Yi Jie Tseng
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
- These authors contributed equally to this work
| | - Sara Soleimani
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
- These authors contributed equally to this work
| | - Tobin Smith
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Maria J Pedroza Romo
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Wisdom Oshireku Abiodun
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Christina M. Egbert
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Deshan Madhusanka
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Derick Bunn
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Bridger Woods
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Evan Tsubaki
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Cameron Stewart
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Seth Brown
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Tzanko Doukov
- Macromolecular Crystallography Group, Structural Molecular Biology Resource, Stanford Synchrotron Radiation Lightsource, Menlo Park, California, United States of America
| | - Joshua L. Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - James D. Moody
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| |
Collapse
|
9
|
Zhou A, Zhang W, Wang B. Host factor TNK2 is required for influenza virus infection. Genes Genomics 2023; 45:771-781. [PMID: 37133719 DOI: 10.1007/s13258-023-01384-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 04/03/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Host factors are required for Influenza virus infection and have great potential to become antiviral target. OBJECTIVE Here we demonstrate the role of TNK2 in influenza virus infection. CRISPR/Cas9 induced TNK2 deletion in A549 cells. METHODS CRISPR/Cas9-mediated deletion of TNK2. Western blotting and qPCR was used to measure the expression of TNK2 and other proteins. RESULTS CRISPR/Cas9-mediated deletion of TNK2 decreased the replication of influenza virus and significantly inhibited the ex-pression of viral proteins and TNK2 inhibitors (XMD8-87 and AIM-100) reduced the expression of influenza M2, while over-expression of TNK2 weakened the resistance of TNK2-knockout cells to influenza virus infection. Furthermore, a decrease of nuclear import of IAV in the infected TNK2 mutant cells was observed in 3 h post-infection. Interestingly, TNK2 deletion enhanced the colocalization of LC3 with autophagic receptor p62 and led to the attenuation of influenza virus-caused accumulation of autophagosomes in TNK2 mutant cells. Further, confocal microscopy visualization result showed that influenza viral matrix 2 (M2) was colocalized with Lamp1 in the infected TNK2 mutant cells in early infection, while almost no colocalization between M2 and Lamp1 was observed in IAV-infected wild-type cells. Moreover, TNK2 depletion also affected the trafficking of early endosome and the movement of influenza viral NP and M2. CONCLUSION Our results identified TNK2 as a critical host factor for influenza viral M2 protein trafficking, suggesting that TNK2 will be an attractive target for the development of antivirals therapeutics.
Collapse
Affiliation(s)
- Ao Zhou
- School of Animal Science and Nutritional Engineering, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Wuhan Polytechnic University, Wuhan, 430023, Hubei, China.
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Hubei Wuhan, Hubei, 430023, China.
| | - Wenhua Zhang
- School of Animal Science and Nutritional Engineering, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Wuhan Polytechnic University, Wuhan, 430023, Hubei, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Hubei Wuhan, Hubei, 430023, China
| | - Baoxin Wang
- School of Animal Science and Nutritional Engineering, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Wuhan Polytechnic University, Wuhan, 430023, Hubei, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Hubei Wuhan, Hubei, 430023, China
| |
Collapse
|
10
|
Kan Y, Paung Y, Seeliger MA, Miller WT. Domain Architecture of the Nonreceptor Tyrosine Kinase Ack1. Cells 2023; 12:900. [PMID: 36980241 PMCID: PMC10047419 DOI: 10.3390/cells12060900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The nonreceptor tyrosine kinase (NRTK) Ack1 comprises a distinct arrangement of non-catalytic modules. Its SH3 domain has a C-terminal to the kinase domain (SH1), in contrast to the typical SH3-SH2-SH1 layout in NRTKs. The Ack1 is the only protein that shares a region of high homology to the tumor suppressor protein Mig6, a modulator of EGFR. The vertebrate Acks make up the only tyrosine kinase (TK) family known to carry a UBA domain. The GTPase binding and SAM domains are also uncommon in the NRTKs. In addition to being a downstream effector of receptor tyrosine kinases (RTKs) and integrins, Ack1 can act as an epigenetic regulator, modulate the degradation of the epidermal growth factor receptor (EGFR), confer drug resistance, and mediate the progression of hormone-sensitive tumors. In this review, we discuss the domain architecture of Ack1 in relation to other protein kinases that possess such defined regulatory domains.
Collapse
Affiliation(s)
- Yagmur Kan
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - YiTing Paung
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Markus A. Seeliger
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
- Department of Veterans Affairs Medical Center, Northport, NY 11768-2200, USA
| |
Collapse
|
11
|
He Q, Qu M, Bao H, Xu Y, Shen T, Tan D, Barkat MQ, Xu C, Zeng LH, Wu X. Multiple post-translational modifications ensure EGFR functionality: Potential therapeutic targets to overcome its drug-resistance mutations. Cytokine Growth Factor Rev 2023; 70:41-53. [PMID: 36934069 DOI: 10.1016/j.cytogfr.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Epidermal growth factor receptor (EGFR) mutation is the most common driver mutation in non-small cell lung cancer (NSCLC). The first-line therapy for advanced NSCLC patients with EGFR-sensitive mutation is the EGFR tyrosine kinase inhibitor (EGFR-TKI). However, most NSCLC patients with EGFR mutation will develop resistant mutations in EGFR-TKI therapy. With further studies, resistance mechanisms represented by EGFR-T790M mutations have revealed the impact of EGFR mutations in situ on EGFR-TKIs sensitivity. The third-generation EGFR-TKIs inhibit both EGFR-sensitive mutations and T790M mutations. The emergence of novel mutations such as EGFR-C797S and EGFR-L718Q may decrease efficacy. Searching for new targets to overcome EGFR-TKI resistance becomes a key challenge. Therefore, an in-depth understanding of the regulatory mechanisms of EGFR is essential to find novel targets to overcome drug-resistant mutations in EGFR-TKIs. EGFR, as a receptor-type tyrosine kinase, undergoes homo/heterodimerization and autophosphorylation upon binding to ligands, which activates multiple downstream signaling pathways. Interestingly, there is growing evidence that the kinase activity of EGFR is affected not only by phosphorylation but also by various post-translational modifications (PTMs, such as S-palmitoylation, S-nitrosylation, Methylation, etc.). In this review, we systematically review the effects of different protein PTMs on EGFR kinase activity and its functionality and suggest that influencing EGFR kinase activity by modulating multiple EGFR sites are potential targets to overcome EGFR-TKIs resistance mutations.
Collapse
Affiliation(s)
- Qiangqiang He
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Meiyu Qu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China
| | - Hangyang Bao
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yana Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tingyu Shen
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dan Tan
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Muhammad Qasim Barkat
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
12
|
He W, Xu L, Ding J, Song L, Yang W, Klooster I, Pilco-Janeta DF, Serrano C, Fang H, Jiang G, Wang X, Yu J, Ou WB. Co-targeting of ACK1 and KIT triggers additive anti-proliferative and -migration effects in imatinib-resistant gastrointestinal stromal tumors. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166690. [PMID: 36921738 DOI: 10.1016/j.bbadis.2023.166690] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023]
Abstract
Most gastrointestinal stromal tumors (GIST) harbor mutated receptor tyrosine kinase (RTK) KIT/PDGFRA, which provides an attractive therapeutic target. However, a majority of GISTs ultimately develop resistance to KIT/PDGFRA inhibitor imatinib, multiple therapeutic targets will be identified as a reasonable strategy in imatinib-resistant GISTs. Biological mechanisms of non-RTK activated CDC42 associated kinase 1 (ACK1) are still unclear, which has been found to be activated in GISTs. In the current report, ACK1 overexpression is demonstrated in GIST cell lines and biopsies. RNA-seq analysis and immunoblotting show that ACK1 expression is dependent on imatinib treatment time in GIST-T1 cell line. The colocalization/complex of KIT and ACK1 in GIST cells are observed, and ACK1 activation is in a partially KIT and CDC42 dependent manner. Treatment with a specific ACK1 inhibitor AIM-100 or ACK1 siRNA, mildly suppresses cell viability, but markedly inhibits cell migration in imatinib sensitive and in imatinib resistant GIST cell lines, which is associated with inactivation of PI3K/AKT/mTOR and RAF/MAPK signaling pathways, and inhibition of epithelial-mesenchymal transition, evidencing upregulation of E-cadherin and downregulation of ZEB1, N-cadherin, vimentin, snail, and/or β-catenin after treatment with AIM-100 or ACK1/CDC42 shRNAs. Combination inhibition of ACK1 and KIT results in additive effects of anti-proliferation and pro-apoptosis as well as cell cycle arrest, and inhibition of invasiveness and migration in vitro and in vivo, compared to either intervention alone through dephosphorylation of KIT downstream intermediates (AKT, S6, and MAPK). Our data suggest that co-targeting of ACK1 and KIT might be a novel therapeutic strategy in imatinib-resistant GIST.
Collapse
Affiliation(s)
- Wangzhen He
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Liangliang Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Jiongyan Ding
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Li Song
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Weili Yang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Isabella Klooster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Daniel F Pilco-Janeta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Sarcoma Translational Research Laboratory, Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | - César Serrano
- Sarcoma Translational Research Laboratory, Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | - Hongming Fang
- Department of Oncology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Guojun Jiang
- Department of Oncology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyan Wang
- Department of Oncology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jiren Yu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Zhu J, Peng Z, Tian X, Wu T, Sun A, Yang W, Lin Q. Activation of E3 ubiquitin ligase WWP2 by non-receptor tyrosine kinase ACK1. IUBMB Life 2023. [PMID: 36773333 DOI: 10.1002/iub.2705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/19/2023] [Indexed: 02/13/2023]
Abstract
WW domain containing E3 ubiquitin protein ligase 2 (WWP2) is a member of the NEDD4 E3 ubiquitin ligase family. WWP2 ligase activity is regulated by the 2, 3-linker auto-inhibition. Tyrosine phosphorylation of the 2, 3-linker was identified as an activating means for releasing the auto-inhibition of WWP2. However, the tyrosine kinase (TK) for the phosphorylation and activation remains unknown. In this report, we have found that non-receptor TK ACK1 binds to the WW3 domain of WWP2 and phosphorylates WWP2. ACK1 phosphorylates WWP2 at the 2, 3-linker and partially activates the ubiquitination ligase activity. Unexpectedly, tyrosine phosphorylation of the 2, 3-linker seems not a major mode for activation of WWP2, as ACK1 causes much higher activation of the 2, 3-linker tyrosine phosphorylation defective mutants of WWP2 than that of wild-type WWP2. Furthermore, epidermal growth factor (EGF) stimulates tyrosine phosphorylation of WWP2 and this EGF-stimulated phosphorylation of WWP2 is mediated by ACK1. Finally, knockdown of WWP2 by shWWP2 inhibits the EGF-dependent cell proliferation of lung cancer A549 cells, suggesting that WWP2 may function in the EGFR signaling in lung cancer progression. Taken together, our findings have revealed a novel mechanism underlying activation of WWP2.
Collapse
Affiliation(s)
- Jun Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ziluo Peng
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xianyan Tian
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tiantian Wu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aiqin Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wannian Yang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
14
|
The noncatalytic regions of the tyrosine kinase Tnk1 are important for activity and substrate specificity. J Biol Chem 2022; 298:102664. [PMID: 36334623 DOI: 10.1016/j.jbc.2022.102664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Human Tnk1 (thirty-eight negative kinase 1) is a member of the Ack family of nonreceptor tyrosine kinases. Tnk1 contains a sterile alpha motif, a tyrosine kinase catalytic domain, an SH3 (Src homology 3) domain, and a large C-terminal region that contains a ubiquitin association domain. However, specific physiological roles for Tnk1 have not been characterized in depth. Here, we expressed and purified Tnk1 from Sf9 insect cells and established an in vitro assay system using a peptide substrate derived from the Wiskott-Aldrich Syndrome Protein (WASP). By Tnk1 expression in mammalian cells, we found that the N-terminal SAM domain is important for self-association and kinase activity. We also studied a fusion protein, originally discovered in a Hodgkin's Lymphoma cell line, that contains an unrelated sequence from the C17ORF61 gene fused to the C-terminus of Tnk1. Cells expressing the fusion protein showed increased tyrosine phosphorylation of cellular substrates relative to cells expressing WT Tnk1. A truncated Tnk1 construct (residues 1-465) also showed enhanced phosphorylation, indicating that the C17ORF61 sequence was dispensable for the effect. Additionally, in vitro kinase assays with the WASP peptide substrate showed no increase in intrinsic Tnk1 activity in C-terminally truncated constructs, suggesting that the truncations did not simply remove an autoinhibitory element. Fluorescence microscopy experiments demonstrated that the C-terminus of Tnk1 plays an important role in the subcellular localization of the kinase. Taken together, our data suggest that the noncatalytic regions of Tnk1 play important roles in governing activity and substrate phosphorylation.
Collapse
|
15
|
Kan Y, Miller WT. Activity of the nonreceptor tyrosine kinase Ack1 is regulated by tyrosine phosphorylation of its Mig6 homology region. FEBS Lett 2022; 596:2808-2820. [PMID: 36178070 PMCID: PMC9879303 DOI: 10.1002/1873-3468.14505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/19/2022] [Indexed: 01/28/2023]
Abstract
Ack1 is a proto-oncogenic tyrosine kinase with homology to the tumour suppressor Mig6, an inhibitor of the epidermal growth factor receptor (EGFR). The residues critical for binding of Mig6 to EGFR are conserved within the Mig6 homology region (MHR) of Ack1. We tested whether intramolecular interactions between the Ack1 MHR and kinase domain (KD) are regulated by phosphorylation. We identified two Src phosphorylation sites within the MHR (Y859, Y860). Addition of Src-phosphorylated MHR to the Ack1 KD enhanced enzymatic activity. Co-expression of Src in cells led to increased Ack1 activity; mutation of Y859/Y860 blocked this increase. Collectively, the data suggest that phosphorylation of the Ack1 MHR regulates its kinase activity. Phosphorylation of Y859/Y860 occurs in cancers of the brain, breast, colon, and prostate, where genomic amplification or somatic mutations of Ack1 play a role in disease progression. Our findings suggest that MHR phosphorylation could contribute to Ack1 dysregulation in tumours.
Collapse
Affiliation(s)
- Yağmur Kan
- Department of Physiology and Biophysics, School of Medicine Stony Brook University NY USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, School of Medicine Stony Brook University NY USA
| |
Collapse
|
16
|
The brain-specific splice variant of the CDC42 GTPase works together with the kinase ACK to downregulate the EGF receptor in promoting neurogenesis. J Biol Chem 2022; 298:102564. [PMID: 36206843 PMCID: PMC9663532 DOI: 10.1016/j.jbc.2022.102564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/12/2022] Open
Abstract
The small GTPase CDC42 plays essential roles in neurogenesis and brain development. Previously, we showed that a CDC42 splice variant that has a ubiquitous tissue distribution specifically stimulates the formation of neural progenitor cells, whereas a brain-specific CDC42 variant, CDC42b, is essential for promoting the transition of neural progenitor cells to neurons. These specific roles of CDC42 and CDC42b in neurogenesis are ascribed to their opposing effects on mTORC1 activity. Specifically, the ubiquitous form of CDC42 stimulates mTORC1 activity and thereby upregulates tissue-specific transcription factors that are essential for neuroprogenitor formation, whereas CDC42b works together with activated CDC42-associated kinase (ACK) to downregulate mTOR expression. Here, we demonstrate that the EGF receptor (EGFR) is an additional and important target of CDC42b and ACK, which is downregulated by their combined actions in promoting neurogenesis. The activation status of the EGFR determines the timing by which neural progenitor cells derived from P19 embryonal carcinoma terminally differentiate into neurons. By promoting EGFR degradation, we found that CDC42b and ACK stimulate autophagy, which protects emerging neurons from apoptosis and helps trigger neural progenitor cells to differentiate into neurons. Moreover, our results reveal that CDC42b is localized in phosphatidylinositol (3,4,5)-triphosphate-enriched microdomains on the plasma membrane, mediated through its polybasic sequence 185KRK187, which is essential for determining its distinct functions. Overall, these findings now highlight a molecular mechanism by which CDC42b and ACK regulate neuronal differentiation and provide new insights into the functional interplay between EGFR degradation and autophagy that occurs during embryonic neurogenesis.
Collapse
|
17
|
Ide M, Tabata N, Yonemura Y, Shirasaki T, Murai K, Wang Y, Ishida A, Okada H, Honda M, Kaneko S, Doi N, Ito S, Yanagawa H. Guanine nucleotide exchange factor DOCK11-binding peptide fused with a single chain antibody inhibits Hepatitis B Virus infection and replication. J Biol Chem 2022; 298:102097. [PMID: 35660020 PMCID: PMC9241042 DOI: 10.1016/j.jbc.2022.102097] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/26/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major global health problem with no established cure. Dedicator of cytokinesis 11 (DOCK11), known as a guanine nucleotide exchange factor (GEF) for Cdc42, is reported to be essential for the maintenance of HBV. However, potential therapeutic strategies targeting DOCK11 have not yet been explored. We have previously developed an in vitro virus method as a more efficient tool for the analysis of proteomics and evolutionary protein engineering. In this study, using the in vitro virus method, we screened and identified a novel antiasialoglycoprotein receptor (ASGR) antibody, ASGR3-10M, and a DOCK11-binding peptide, DCS8-42A, for potential use in HBV infection. We further constructed a fusion protein (10M-D42AN) consisting of ASGR3-10M, DCS8-42A, a fusogenic peptide, and a nuclear localization signal to deliver the peptide inside hepatocytes. We show using immunofluorescence staining that 10M-D42AN was endocytosed into early endosomes and released into the cytoplasm and nucleus. Since DCS8-42A shares homology with activated cdc42-associated kinase 1 (Ack1), which promotes EGFR endocytosis required for HBV infection, we also found that 10M-D42AN inhibited endocytosis of EGFR and Ack1. Furthermore, we show 10M-D42AN suppressed the function of DOCK11 in the host DNA repair system required for covalently closed circular DNA synthesis and suppressed HBV proliferation in mice. In conclusion, this study realizes a novel hepatocyte-specific drug delivery system using an anti-ASGR antibody, a fusogenic peptide, and DOCK11-binding peptide to provide a novel treatment for HBV.
Collapse
Affiliation(s)
- Mayuko Ide
- Research Department, Purotech Bio Inc, Yokohama, Kanagawa, 230-0045, Japan
| | - Noriko Tabata
- Research Department, Purotech Bio Inc, Yokohama, Kanagawa, 230-0045, Japan
| | - Yuko Yonemura
- Research Department, Purotech Bio Inc, Yokohama, Kanagawa, 230-0045, Japan
| | - Takayoshi Shirasaki
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Health Medicine, Kanazawa, Ishikawa, 920-0942, Japan
| | - Kazuhisa Murai
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Health Medicine, Kanazawa, Ishikawa, 920-0942, Japan
| | - Ying Wang
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Health Medicine, Kanazawa, Ishikawa, 920-0942, Japan
| | - Atsuya Ishida
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Health Medicine, Kanazawa, Ishikawa, 920-0942, Japan
| | - Hikari Okada
- Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Ishikawa, 920-8641, Japan
| | - Masao Honda
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Health Medicine, Kanazawa, Ishikawa, 920-0942, Japan; Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Ishikawa, 920-8641, Japan
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Ishikawa, 920-8641, Japan
| | - Nobuhide Doi
- Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa, 223-8522, Japan
| | - Satoru Ito
- Research Department, Purotech Bio Inc, Yokohama, Kanagawa, 230-0045, Japan
| | - Hiroshi Yanagawa
- Research Department, Purotech Bio Inc, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
18
|
Clayton NS, Fox M, Vicenté-Garcia JJ, Schroeder CM, Littlewood TD, Wilde JI, Krishnan K, Brown MJB, Crafter C, Mott HR, Owen D. Assembly of nuclear dimers of PI3K regulatory subunits is regulated by the Cdc42-activated tyrosine kinase ACK. J Biol Chem 2022; 298:101916. [PMID: 35429500 PMCID: PMC9127371 DOI: 10.1016/j.jbc.2022.101916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Activated Cdc42-associated kinase (ACK) is an oncogenic nonreceptor tyrosine kinase associated with poor prognosis in several human cancers. ACK promotes proliferation, in part by contributing to the activation of Akt, the major effector of class 1A phosphoinositide 3-kinases (PI3Ks), which transduce signals via membrane phosphoinositol lipids. We now show that ACK also interacts with other key components of class 1A PI3K signaling, the PI3K regulatory subunits. We demonstrate ACK binds to all five PI3K regulatory subunit isoforms and directly phosphorylates p85α, p85β, p50α, and p55α on Tyr607 (or analogous residues). We found that phosphorylation of p85β promotes cell proliferation in HEK293T cells. We demonstrate that ACK interacts with p85α exclusively in nuclear-enriched cell fractions, where p85α phosphorylated at Tyr607 (pTyr607) also resides, and identify an interaction between pTyr607 and the N-terminal SH2 domain that supports dimerization of the regulatory subunits. We infer from this that ACK targets p110-independent p85 and further postulate that these regulatory subunit dimers undertake novel nuclear functions underpinning ACK activity. We conclude that these dimers represent a previously undescribed mode of regulation for the class1A PI3K regulatory subunits and potentially reveal additional avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Natasha S Clayton
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Millie Fox
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Trevor D Littlewood
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Jonathon I Wilde
- GlaxoSmithKline Medicines Research Centre, Screening and Compound Profiling, Stevenage, Herts, United Kingdom
| | - Kadalmani Krishnan
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Murray J B Brown
- GlaxoSmithKline Medicines Research Centre, Screening and Compound Profiling, Stevenage, Herts, United Kingdom
| | - Claire Crafter
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Helen R Mott
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| | - Darerca Owen
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
19
|
Miller KJ, Asim M. Unravelling the Role of Kinases That Underpin Androgen Signalling in Prostate Cancer. Cells 2022; 11:cells11060952. [PMID: 35326402 PMCID: PMC8946764 DOI: 10.3390/cells11060952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The androgen receptor (AR) signalling pathway is the key driver in most prostate cancers (PCa), and is underpinned by several kinases both upstream and downstream of the AR. Many popular therapies for PCa that target the AR directly, however, have been circumvented by AR mutation, such as androgen receptor variants. Some upstream kinases promote AR signalling, including those which phosphorylate the AR and others that are AR-regulated, and androgen regulated kinase that can also form feed-forward activation circuits to promotes AR function. All of these kinases represent potentially druggable targets for PCa. There has generally been a divide in reviews reporting on pathways upstream of the AR and those reporting on AR-regulated genes despite the overlap that constitutes the promotion of AR signalling and PCa progression. In this review, we aim to elucidate which kinases—both upstream and AR-regulated—may be therapeutic targets and require future investigation and ongoing trials in developing kinase inhibitors for PCa.
Collapse
|
20
|
Lee HW, Choi Y, Lee AR, Yoon CH, Kim KH, Choi BS, Park YK. Hepatocyte Growth Factor-Dependent Antiviral Activity of Activated cdc42-Associated Kinase 1 Against Hepatitis B Virus. Front Microbiol 2022; 12:800935. [PMID: 35003030 PMCID: PMC8733702 DOI: 10.3389/fmicb.2021.800935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/06/2021] [Indexed: 01/04/2023] Open
Abstract
Activated cdc42-associated kinase 1 (ACK1) is a well-known non-receptor tyrosine kinase that regulates cell proliferation and growth through activation of cellular signaling pathways, including mitogen-activated protein kinase (MAPK). However, the anti-HBV activity of ACK1 has not been elucidated. This study aimed to investigate the role of ACK1 in the HBV life cycle and the mechanism underlying the anti-HBV activity of ACK1. To examine the antiviral activity of ACK1, we established HepG2-ACK1 cells stably overexpressing ACK1. The HBV life cycle, including HBeAg/HBsAg secretion, HBV DNA/transcription, and enhancer activity, was analyzed in HepG2 and HepG2-ACK1 cells with HBV replication-competent HBV 1.2mer (HBV 1.2). Finally, the anti-HBV activity of ACK1 was examined in an HBV infection system. ACK1 suppressed HBV gene expression and transcription in HepG2 and HepG2-ACK1 cells. Furthermore, ACK1 inhibited HBV replication by decreasing viral enhancer activity. ACK1 exhibited its anti-HBV activity via activation of Erk1/2, which consequently downregulated the expression of HNF4α binding to HBV enhancers. Furthermore, hepatocyte growth factor (HGF) induced ACK1 expression at an early stage. Finally, ACK1 mediated the antiviral effect of HGF in the HBV infection system. These results indicated that ACK1 induced by HGF inhibited HBV replication at the transcriptional level by activating the MAPK-HNF signaling pathway. Our findings suggest that ACK1 is a potentially novel upstream molecule of MAPK-mediated anti-HBV activity.
Collapse
Affiliation(s)
- Hye Won Lee
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Cheongju, South Korea
| | - Yongwook Choi
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Cheongju, South Korea
| | - Ah Ram Lee
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, South Korea
| | - Cheol-Hee Yoon
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Cheongju, South Korea
| | - Kyun-Hwan Kim
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, South Korea
| | - Byeong-Sun Choi
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Cheongju, South Korea
| | - Yong Kwang Park
- Division of Chronic Viral Diseases, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Cheongju, South Korea
| |
Collapse
|
21
|
TNK1 is a ubiquitin-binding and 14-3-3-regulated kinase that can be targeted to block tumor growth. Nat Commun 2021; 12:5337. [PMID: 34504101 PMCID: PMC8429728 DOI: 10.1038/s41467-021-25622-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/20/2021] [Indexed: 02/08/2023] Open
Abstract
TNK1 is a non-receptor tyrosine kinase with poorly understood biological function and regulation. Here, we identify TNK1 dependencies in primary human cancers. We also discover a MARK-mediated phosphorylation on TNK1 at S502 that promotes an interaction between TNK1 and 14-3-3, which sequesters TNK1 and inhibits its kinase activity. Conversely, the release of TNK1 from 14-3-3 allows TNK1 to cluster in ubiquitin-rich puncta and become active. Active TNK1 induces growth factor-independent proliferation of lymphoid cells in cell culture and mouse models. One unusual feature of TNK1 is a ubiquitin-association domain (UBA) on its C-terminus. Here, we characterize the TNK1 UBA, which has high affinity for poly-ubiquitin. Point mutations that disrupt ubiquitin binding inhibit TNK1 activity. These data suggest a mechanism in which TNK1 toggles between 14-3-3-bound (inactive) and ubiquitin-bound (active) states. Finally, we identify a TNK1 inhibitor, TP-5801, which shows nanomolar potency against TNK1-transformed cells and suppresses tumor growth in vivo.
Collapse
|
22
|
Liu J, Yang L, He A, Ke M, Fu C, Gao W, Xu R, Tian R. Stable and EGF-Induced Temporal Interactome Profiling of CBL and CBLB Highlights Their Signaling Complex Diversity. J Proteome Res 2021; 20:3709-3719. [PMID: 34134489 DOI: 10.1021/acs.jproteome.1c00284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The epidermal growth factor receptor (EGFR) signal modulates cell proliferation, migration, and survival. Aberrant activation of EGFR constitutes the major cause of various cancers. Receptor ubiquitination and degradation mediated by CBL proteins play negative regulatory roles and control the intensity and duration of the signaling. With the construction of stable cell lines inducibly expressing FLAG-tagged CBL or CBLB, we identified 102 and 82 stable interacting proteins of CBL and CBLB, respectively, through the affinity purification followed by mass spectrometry (AP-MS) approach. Time-resolved profiling at six different time points combined with functional annotations of the temporal interactomes provides insights into the dynamic assembly of signal proteins upon EGFR signaling activation. Comparison between the interactomes of CBL and CBLB indicates their redundant but also complementary functions. Importantly, we validated the stable association of EPS15L1 and ITSN2 and temporal association of TNK2 to both CBL and CBLB through biochemical assays. Collectively, these results offer a useful resource for CBL and CBLB interactomes and highlight their prominent and diverse roles in the EGFR signaling network.
Collapse
Affiliation(s)
- Jie Liu
- Department of Oncology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China.,The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Lijun Yang
- Department of Oncology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China.,The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - An He
- Department of Chemistry, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mi Ke
- Department of Chemistry, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Changying Fu
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weina Gao
- Department of Chemistry, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruilian Xu
- Department of Oncology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China.,The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Ruijun Tian
- Department of Chemistry, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
23
|
Zhang A, Zhang R, Yang Z, Tian R. TNK2 promoted esophageal cancer progression via activating egfr-akt signaling. J Clin Lab Anal 2021; 35:e23700. [PMID: 33484472 PMCID: PMC7958005 DOI: 10.1002/jcla.23700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Background This study investigated the clinical implication of TNK2 expression in esophageal cancer patients’ cancer tissue samples. Methods The expression of TNK2 in esophageal cancer tissues and para‐carcinoma tissue was assessed with immunohistochemistry and Western blot analysis; besides, the proteins of CDC42, EGFR, and Akt were also analyzed. Then, Kaplan‐Meier survival curves of TNK2 protein expression level were assayed with 184 esophageal cancer patients from TCGA database. Moreover, with multiple linear regression analysis, we detected the correlations of TNK2 expression associated with tumor differentiation degree and metastasis status. Results It revealed that TNK2 was highly expressed in the cytoplasm of esophageal cancer tissues compared with para‐carcinoma tissue; besides, the proteins of CDC42, EGFR, and Akt were also up‐regulated in different levels of esophageal cancer tissues. However, there was no significant difference of the overall survival time of TNK2 protein expression in 184 esophageal cancer patients from TCGA database (p = 0.37). But, in the included study samples of our study, there was positive coefficience between TNK2 protein expression and differentiation degree in esophageal cancer with multiple linear regression analysis [R = 0.928, 95% confidence interval (0.085‐0.12)]. Conclusion Our results indicated that TNK2 was a potential diagnostic marker and promoted esophageal cancer progression through activating EGFR‐AKT signaling.
Collapse
Affiliation(s)
- Anqing Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Rongxin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Zhiming Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Rui Tian
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| |
Collapse
|
24
|
Zhu J, Liu Y, Zhao M, Cao K, Ma J, Peng S. Identification of downstream signaling cascades of ACK1 and prognostic classifiers in non-small cell lung cancer. Aging (Albany NY) 2021; 13:4482-4502. [PMID: 33495411 PMCID: PMC7906148 DOI: 10.18632/aging.202408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/27/2020] [Indexed: 12/29/2022]
Abstract
Activated Cdc42-associated kinase 1 (ACK1) is an oncogene in multiple cancers, but the underlying mechanisms of its oncogenic role remain unclear in non-small cell lung cancer (NSCLC). Herein, we comprehensively investigated the ACK1-regulated cell processes and downstream signaling pathways, as well as its prognostic value in NSCLC. We found that ACK1 gene amplification was associated with mRNA levels in The Cancer Genome Atlas (TCGA) lung cancer cohort. The Oncomine databases showed significantly elevated ACK1 levels in lung cancer. In vitro, an ACK1 inhibitor (dasatinib) increased the sensitivity of NSCLC cell lines to AKT or MEK inhibitors. RNA-sequencing results demonstrated that an ACK1 deficiency in A549 cells affected the MAPK, PI3K/AKT, and Wnt pathways. These results were validated by gene set enrichment analysis (GSEA) of data from 188 lung cancer cell lines. Using Cytoscape, we dissected 14 critical ACK1-regulated genes. The signature with the 14 genes and ACK1 could significantly dichotomize the TCGA lung cohort regarding overall survival. The prognostic accuracy of this signature was confirmed in five independent lung cancer cohorts and was further validated by a prognostic nomogram. Our study unveiled several downstream signaling pathways for ACK1, and the proposed signature may be a promising prognostic predictor for NSCLC.
Collapse
Affiliation(s)
- Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Yang Liu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Meng Zhao
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Kui Cao
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Shiyun Peng
- Department of Precision Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| |
Collapse
|
25
|
Medda N, De SK, Maiti S. Different mechanisms of arsenic related signaling in cellular proliferation, apoptosis and neo-plastic transformation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111752. [PMID: 33396077 DOI: 10.1016/j.ecoenv.2020.111752] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/12/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023]
Abstract
Arsenic is a toxic heavy metal vastly dispersed all over the earth crust. It manifests several major adverse health issues to millions of arsenic exposed populations. Arsenic is associated with different types of cancer, cardiovascular disorders, diabetes, hypertension and many other diseases. On the contrary, arsenic (arsenic trioxide, As2O3) is used as a chemotherapeutic agent in the treatment of acute promyelocytic leukemia. Balance between arsenic induced cellular proliferations and apoptosis finally decide the outcome of its transformation rate. Arsenic propagates signals via cellular and nuclear pathways depending upon the chemical nature, and metabolic-fates of the arsenical compounds. Arsenic toxicity is propagated via ROS induced stress to DNA-repair mechanism and mitochondrial stability in the cell. ROS induced alteration in p53 regulation and some mitogen/ oncogenic functions determine the transformation outcome influencing cyclin-cdk complexes. Growth factor regulator proteins such as c-Jun, c-fos and c-myc are influenced by chronic arsenic exposure. In this review we have delineated arsenic induced ROS regulations of epidermal growth factor receptor (EGFR), NF-ĸβ, MAP kinase, matrix-metalloproteinases (MMPs). The role of these signaling molecules has been discussed in relation to cellular apoptosis, cellular proliferation and neoplastic transformation. The arsenic stimulated pathways which help in proliferation and neoplastic transformation ultimately resulted in cancer manifestation whereas apoptotic pathways inhibited carcinogenesis. Therapeutic strategies against arsenic should be designed taking into account all these factors.
Collapse
Affiliation(s)
- Nandita Medda
- Center for Life Sciences, Vidyasagar University, Midnapore-721102, West Bengal, India; Post Graduate Department of Biochemistry and Biotechnology Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Midnapore-721102, West Bengal, India
| | - Subrata Kumar De
- Professor, Dept. of Zoology, Vidyasagar University, Midnapore, 721102, West Bengal, India; (on lien) Vice Chancellor, Mahatma Gandhi University, Purba Medinipur, 721628, West Bengal, India.
| | - Smarajit Maiti
- Post Graduate Department of Biochemistry and Biotechnology Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Midnapore-721102, West Bengal, India.
| |
Collapse
|
26
|
Ji Z, Njauw CN, Guhan S, Kumar R, Reddy B, Rajadurai A, Flaherty K, Tsao H. Loss of ACK1 Upregulates EGFR and Mediates Resistance to BRAF Inhibition. J Invest Dermatol 2020; 141:1317-1324.e1. [PMID: 33159968 DOI: 10.1016/j.jid.2020.06.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/27/2020] [Accepted: 06/29/2020] [Indexed: 10/23/2022]
Abstract
Targeted BRAF(V600E) suppression by selective BRAF inhibitors (BRAFis; e.g., vemurafenib and dabrafenib) has led to a sea change in the treatment of metastatic melanoma. Despite frequent upfront responses, acquired resistance has compromised long-term applicability. Among the various mechanisms of resistance, activation of multiple receptor tyrosine kinases is a known critical factor that contributes to vemurafenib resistance. EGFR activation has been recurrently identified in a set of vemurafenib-resistant melanomas, but little is known about how EGFR, or possibly other receptor tyrosine kinases, becomes activated. Here, we report that ACK1, a protein kinase that modulates EGFR turnover, is downregulated in vemurafenib-resistant melanoma cells. We also found that ACK1 depletion with short hairpin RNA decreased EGFR degradation when activated by epidermal growth factor, increased EGFR protein expression, and conferred resistance to BRAFis both in vitro and in vivo. Vemurafenib resistance mediated by ACK1 inhibition can be reversed by the EGFR inhibitor gefitinib. Our data indicate that ACK1 loss may be a post-transcriptional mechanism that increases EGFR signaling and contributes to drug resistance.
Collapse
Affiliation(s)
- Zhenyu Ji
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ching-Ni Njauw
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samantha Guhan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Raj Kumar
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bobby Reddy
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anpuchelvi Rajadurai
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Flaherty
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
27
|
Gu J, Qian L, Zhang G, Mahajan NP, Owonikoko TK, Ramalingam SS, Sun SY. Inhibition of ACK1 delays and overcomes acquired resistance of EGFR mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib. Lung Cancer 2020; 150:26-35. [PMID: 33049499 DOI: 10.1016/j.lungcan.2020.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVES The emergence of acquired resistance to the third generation EGFR inhibitor, osimertinib (AZD9291 or TAGRISSO™), is an unavoidable huge clinical challenge. The involvement of ACK1, a non-receptor tyrosine kinase with an oncogenic function, in regulating cell response to osimertinib has not been investigated and thus is the focus of this study. MATERIAL AND METHODS Drug effects on cell growth were evaluated by measuring cell numbers and colony formation. Apoptosis was monitored with flow cytometry for annexin V-positive cells and Western blotting for protein cleavage. Intracellular protein and mRNA alterations were detected with Western blotting and qRT-PCR, respectively. Drug effects on delaying osimertinib acquired resistance were determined using colony formation in vitro and xenografts in nude mice in vivo, respectively. Cell senescence was assayed by β-galactosidase staining. RESULTS Inhibition of ACK1 with the novel ACK1 inhibitor, (R)-9b synergized with osimertinib in inhibiting the growth of EGFR mutant NSCLC cell lines. Similar results were also generated with ACK1 gene knockdown. The combination of osimertinib and (R)-9b enhanced induction of apoptosis. In both in vitro and in vivo long-term resistance delay assays, the combination of (R)-9b and osimertinib clearly delayed the emergence of osimertinib-resistance. Further, the (R)-9b and osimertinib combination was also effective in inhibiting the growth of EGFR mutant NSCLC cell lines with acquired resistance to osimertinib, which possess elevated levels of ACK1, and the growth of osimertinib-resistant tumors in vivo. In some resistant cell lines, the combinations induced senescence in addition to induction of apoptosis. CONCLUSIONS These novel findings suggest that ACK1 inhibition might be a potential and innovative strategy for delaying and overcoming osimertinb acquired resistance.
Collapse
Affiliation(s)
- Jiajia Gu
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, Jiangsu, PR China; Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Luxi Qian
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, Jiangsu, PR China; Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Guojing Zhang
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Nupam P Mahajan
- Department of Surgery, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Taofeek K Owonikoko
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Suresh S Ramalingam
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Shi-Yong Sun
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
28
|
Wang B, Song K, Chen L, Su H, Gao L, Liu J, Huang A. Targeted inhibition of ACK1 can inhibit the proliferation of hepatocellular carcinoma cells through the PTEN/AKT/mTOR pathway. Cell Biochem Funct 2020; 38:642-650. [PMID: 32162707 DOI: 10.1002/cbf.3522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/23/2020] [Accepted: 02/24/2020] [Indexed: 01/04/2023]
Abstract
Activated Cdc42-associated kinase 1 (ACK1) expression is upregulated in hepatocellular carcinoma (HCC) tissues and other tumour tissues. However, the function and regulatory mechanism of ACK1 in HCC remains unclear. In this study, the expression of pTyr284-ACK1, pSer473-AKT and PTEN in HCC was detected by immunohistochemistry, and its clinicopathological significance was analysed. Then, ACK1-targeted small molecule inhibitors AIM-100 and Dasatinib were used to treat cells SK-Hep-1 and HepG2, and changes in activity and biological behaviours of PTEN/AKT/mTOR signalling pathway were observed. The results showed that pTyr284-ACK1 protein was highly expressed in HCC tissues and was related to the poor prognosis of patients; the expression of pTyr284-ACK1 protein was positively correlated with pSer473-AKT and negatively correlated with PTEN. In addition, after treatment either with AIM-100 or Dasatinib, both proliferation of two cells and migration, invasion of SK-Hep-1 cells were all significantly inhibited. Meanwhile, ACK1, pTyr284-ACK1, pSer473-AKT, mTOR and EGFR were down-regulated; PTEN was up-regulated when analysed by western-blot in SK-Hep-1 cells. These results demonstrated that ACK1 may promote HCC development via PTEN/AKT/mTOR pathway. Targeted inhibition of ACK1 may be a novel therapeutic strategy for HCC. SIGNIFICANCE OF THE STUDY: Hepatocellular carcinoma (HCC) is a common malignant tumour with high mortality. Our study showed that ACK1 and pTyr284-ACK1 are highly expressed in HCC and may promote HCC development through the PTEN/AKT/mTOR signalling pathway. Targeted inhibition of ACK1 expression with small inhibitors AIM-100 and Dasatinib may weaken tumour cells ability of proliferation, migration and invasion. Our results suggested that downregulation of ACK1 may be a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Bin Wang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai Song
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lihong Chen
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Hongying Su
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Lingyun Gao
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Aimin Huang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
29
|
The non-receptor tyrosine kinase ACK: regulatory mechanisms, signalling pathways and opportunities for attACKing cancer. Biochem Soc Trans 2020; 47:1715-1731. [PMID: 31845724 DOI: 10.1042/bst20190176] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/30/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022]
Abstract
Activated Cdc42-associated kinase or ACK, is a non-receptor tyrosine kinase and an effector protein for the small G protein Cdc42. A substantial body of evidence has accumulated in the past few years heavily implicating ACK as a driver of oncogenic processes. Concomitantly, more is also being revealed regarding the signalling pathways involving ACK and molecular details of its modes of action. Some details are also available regarding the regulatory mechanisms of this kinase, including activation and regulation of its catalytic activity, however, a full understanding of these aspects remains elusive. This review considers the current knowledge base concerning ACK and summarizes efforts and future prospects to target ACK therapeutically in cancer.
Collapse
|
30
|
Endo M, Druso JE, Cerione RA. The two splice variant forms of Cdc42 exert distinct and essential functions in neurogenesis. J Biol Chem 2020; 295:4498-4512. [PMID: 32071086 DOI: 10.1074/jbc.ra119.011837] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/31/2020] [Indexed: 12/29/2022] Open
Abstract
The small GTPase cell division cycle 42 (CDC42) plays essential roles in neurogenesis and brain development. Previously, using murine embryonic P19 cells as a model system, we showed that CDC42 stimulates mTOR complex 1 (mTORC1) activity and thereby up-regulates transcription factors required for the formation of neural progenitor cells. However, paradoxically, although endogenous CDC42 is required for both the initial transition of undifferentiated P19 cells to neural progenitors and their ultimate terminal differentiation into neurons, ectopic CDC42 overexpression promotes only the first stage of neurogenesis (i.e. the formation of neuroprogenitors) and not the second phase (differentiation into neurons). Here, using both P19 cells and mouse embryonic stem cells, we resolve this paradox, demonstrating that two splice variants of CDC42, differing only in nine amino acid residues in their very C-terminal regions, play distinct roles in neurogenesis. We found that a CDC42 splice variant that has a ubiquitous tissue distribution, termed here as CDC42u, specifically drives the formation of neuroprogenitor cells, whereas a brain-specific CDC42 variant, CDC42b, is essential for promoting the transition of neuroprogenitor cells to neurons. We further show that the specific roles of CDC42u and CDC42b in neurogenesis are due to their opposing effects on mTORC1 activity. Specifically, CDC42u stimulated mTORC1 activity and thereby induced neuroprogenitor formation, whereas CDC42b worked together with activated CDC42-associated kinase (ACK) in down-regulating mTOR expression and promoting neuronal differentiation. These findings highlight the remarkable functional specificities of two highly similar CDC42 splice variants in regulating distinct stages of neurogenesis.
Collapse
Affiliation(s)
- Makoto Endo
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853
| | - Joseph E Druso
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853
| | - Richard A Cerione
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853 .,Department of Chemistry and Chemical Biology, Baker Laboratory, Cornell University, Ithaca, New York 14853
| |
Collapse
|
31
|
Research Progress of the Functional Role of ACK1 in Breast Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1018034. [PMID: 31772931 PMCID: PMC6854235 DOI: 10.1155/2019/1018034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/13/2019] [Indexed: 01/25/2023]
Abstract
ACK1 is a nonreceptor tyrosine kinase with a unique structure, which is tightly related to the biological behavior of tumors. Previous studies have demonstrated that ACK1 was involved with multiple signaling pathways of tumor progression. Its crucial role in tumor cell proliferation, apoptosis, invasion, and metastasis was tightly related to the prognosis and clinicopathology of cancer. ACK1 has a unique way of regulating cellular pathways, different from other nonreceptor tyrosine kinases. As an oncogenic kinase, recent studies have shown that ACK1 plays a critical regulatory role in the initiation and progression of tumors. In this review, we will be summarizing the structural characteristics, activation, and regulation of ACK1 in breast cancer, aiming to deeply understand the functional and mechanistic role of ACK1 and provide novel therapeutic strategies for breast cancer treatment.
Collapse
|
32
|
Jenkins C, Luty SB, Maxson JE, Eide CA, Abel ML, Togiai C, Nemecek ER, Bottomly D, McWeeney SK, Wilmot B, Loriaux M, Chang BH, Tyner JW. Synthetic lethality of TNK2 inhibition in PTPN11-mutant leukemia. Sci Signal 2018; 11:eaao5617. [PMID: 30018082 PMCID: PMC6168748 DOI: 10.1126/scisignal.aao5617] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The protein tyrosine phosphatase PTPN11 is implicated in the pathogenesis of juvenile myelomonocytic leukemia (JMML), acute myeloid leukemia (AML), and other malignancies. Activating mutations in PTPN11 increase downstream proliferative signaling and cell survival. We investigated the signaling upstream of PTPN11 in JMML and AML cells and found that PTPN11 was activated by the nonreceptor tyrosine/serine/threonine kinase TNK2 and that PTPN11-mutant JMML and AML cells were sensitive to TNK2 inhibition. In cultured human cell-based assays, PTPN11 and TNK2 interacted directly, enabling TNK2 to phosphorylate PTPN11, which subsequently dephosphorylated TNK2 in a negative feedback loop. Mutations in PTPN11 did not affect this physical interaction but increased the basal activity of PTPN11 such that TNK2-mediated activation was additive. Consequently, coexpression of TNK2 and mutant PTPN11 synergistically increased mitogen-activated protein kinase (MAPK) signaling and enhanced colony formation in bone marrow cells from mice. Chemical inhibition of TNK2 blocked MAPK signaling and colony formation in vitro and decreased disease burden in a patient with PTPN11-mutant JMML who was treated with the multikinase (including TNK2) inhibitor dasatinib. Together, these data suggest that TNK2 is a promising therapeutic target for PTPN11-mutant leukemias.
Collapse
MESH Headings
- Animals
- Child
- Dasatinib/pharmacology
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelomonocytic, Juvenile/drug therapy
- Leukemia, Myelomonocytic, Juvenile/enzymology
- Leukemia, Myelomonocytic, Juvenile/genetics
- Leukemia, Myelomonocytic, Juvenile/pathology
- Male
- Mice
- Prognosis
- Protein Kinase Inhibitors/pharmacology
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Signal Transduction
- Survival Rate
- Synthetic Lethal Mutations
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- Chelsea Jenkins
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samuel B Luty
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Julia E Maxson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher A Eide
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Melissa L Abel
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Corinne Togiai
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Eneida R Nemecek
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Daniel Bottomly
- Oregon Clinical and Translational Research Institute, Portland, OR 97239, USA
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Oregon Clinical and Translational Research Institute, Portland, OR 97239, USA
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Beth Wilmot
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Oregon Clinical and Translational Research Institute, Portland, OR 97239, USA
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Marc Loriaux
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Bill H Chang
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA.
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jeffrey W Tyner
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
33
|
Shao G, Wang R, Sun A, Wei J, Peng K, Dai Q, Yang W, Lin Q. The E3 ubiquitin ligase NEDD4 mediates cell migration signaling of EGFR in lung cancer cells. Mol Cancer 2018; 17:24. [PMID: 29455656 PMCID: PMC5817799 DOI: 10.1186/s12943-018-0784-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/01/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND EGFR-dependent cell migration plays an important role in lung cancer progression. Our previous study observed that the HECT E3 ubiquitin ligase NEDD4 is significantly correlated with tumor metastasis and required for migration and invasion signaling of EGFR in gastric cancer cells. However, how NEDD4 promotes the EGFR-dependent lung cancer cell migration is unknown. This study is to elucidate the mechanism by which NEDD4 mediates the EGFR lung cancer migration signaling. METHODS Lentiviral vector-loaded NEDD4 shRNA was used to deplete endogenous NEDD4 in lung cancer cell lines. Effects of the NEDD4 knockdown on the EGFR-dependent or independent lung cancer cell migration were determined using the wound-healing and transwell assays. Association of NEDD4 with activated EGFR was assayed by co-immunoprecipitation. Co-expression of NEDD4 with EGFR or PTEN was determined by immunohistochemical (IHC) staining in 63 lung adenocarcinoma tissue samples. Effects of NEDD4 ectopic expression or knockdown on PTEN ubiquitination and down-regulation, AKT activation and lysosomal secretion were examined using the GST-Uba pulldown assay, immunoblotting, immunofluorescent staining and a human cathepsin B ELISA assay respectively. The specific cathepsin B inhibitor CA-074Me was used for assessing the role of cathepsin B in lung cancer cell migration. RESULTS Knockdown of NEDD4 significantly reduced EGF-stimulated cell migration in non-small cell lung carcinoma (NSCLC) cells. Co-immunoprecipitation assay found that NEDD4 is associated with EGFR complex upon EGF stimulation, and IHC staining indicates that NEDD4 is co-expressed with EGFR in lung adenocarcinoma tumor tissues, suggesting that NEDD4 might mediate lung cancer cell migration by interaction with the EGFR signaling complex. Interestingly, NEDD4 promotes the EGF-induced cathepsin B secretion, possibly through lysosomal exocytosis, as overexpression of the ligase-dead mutant of NEDD4 impedes lysosomal secretion, and knockdown of NEDD4 significantly reduced extracellular amount of cathepsin B induced by EGF. Consistent with the role of NEDD4, cathepsin B is pivotal for both basal and the EGF-stimulated lung cancer cell migration. Our studies propose a novel mechanism underlying the EGFR-promoted lung cancer cell migration that is mediated by NEDD4 through regulation of cathepsin B secretion. CONCLUSION NEDD4 mediates the EGFR lung cancer cell migration signaling through promoting lysosomal secretion of cathepsin B.
Collapse
Affiliation(s)
- Genbao Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Ranran Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Aiqin Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jing Wei
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Ke Peng
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qian Dai
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Wannian Yang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
34
|
Liu L, Thaker TM, Freed DM, Frazier N, Malhotra K, Lemmon MA, Jura N. Regulation of Kinase Activity in the Caenorhabditis elegans EGF Receptor, LET-23. Structure 2018; 26:270-281.e4. [PMID: 29358026 DOI: 10.1016/j.str.2017.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/07/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
Abstract
In the active HER receptor dimers, kinases play distinct roles; one is the catalytically active kinase and the other is its allosteric activator. This specialization enables signaling by the catalytically inactive HER3, which functions exclusively as an allosteric activator upon heterodimerization with other HER receptors. It is unclear whether the allosteric activation mechanism evolved before HER receptors functionally specialized. We determined the crystal structure of the kinase domain of the only EGF receptor in Caenorhabditis elegans, LET-23. Our structure of a non-human EGFR kinase reveals autoinhibitory features conserved in the human counterpart. Strikingly, mutations within the putative allosteric dimer interface abrogate activity of the isolated LET-23 kinase and of the full-length receptor despite these regions being only partially conserved with human EGFR. Our results indicate that ancestral EGFRs have built-in features that poise them for allosteric activation that could facilitate emergence of the catalytically dead, yet functional, orthologs.
Collapse
Affiliation(s)
- Lijun Liu
- Cardiovascular Research Institute, University of California - San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California - San Francisco, San Francisco, CA 94158, USA
| | - Tarjani M Thaker
- Cardiovascular Research Institute, University of California - San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California - San Francisco, San Francisco, CA 94158, USA
| | - Daniel M Freed
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Nicole Frazier
- Cardiovascular Research Institute, University of California - San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California - San Francisco, San Francisco, CA 94158, USA
| | - Ketan Malhotra
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Mark A Lemmon
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California - San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California - San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
35
|
Caldieri G, Malabarba MG, Di Fiore PP, Sigismund S. EGFR Trafficking in Physiology and Cancer. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:235-272. [PMID: 30097778 DOI: 10.1007/978-3-319-96704-2_9] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling from the epidermal growth factor receptor (EGFR) elicits multiple biological responses, including cell proliferation, migration, and survival. Receptor endocytosis and trafficking are critical physiological processes that control the strength, duration, diversification, and spatial restriction of EGFR signaling through multiple mechanisms, which we review in this chapter. These mechanisms include: (i) regulation of receptor density and activation at the cell surface; (ii) concentration of receptors into distinct nascent endocytic structures; (iii) commitment of the receptor to different endocytic routes; (iv) endosomal sorting and postendocytic trafficking of the receptor through distinct pathways, and (v) recycling to restricted regions of the cell surface. We also highlight how communication between organelles controls EGFR activity along the endocytic route. Finally, we illustrate how abnormal trafficking of EGFR oncogenic mutants, as well as alterations of the endocytic machinery, contributes to aberrant EGFR signaling in cancer.
Collapse
Affiliation(s)
- Giusi Caldieri
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy
| | - Maria Grazia Malabarba
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy
| | - Pier Paolo Di Fiore
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy
| | - Sara Sigismund
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via Santa Sofia 9/1, 20122, Milan, Italy.
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141, Milan, Italy.
| |
Collapse
|
36
|
Del Mar Masdeu M, Armendáriz BG, Torre AL, Soriano E, Burgaya F, Ureña JM. Identification of novel Ack1-interacting proteins and Ack1 phosphorylated sites in mouse brain by mass spectrometry. Oncotarget 2017; 8:101146-101157. [PMID: 29254152 PMCID: PMC5731862 DOI: 10.18632/oncotarget.20929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/26/2017] [Indexed: 12/04/2022] Open
Abstract
Ack1 (activated Cdc42-associated tyrosine kinase) is a non-receptor tyrosine kinase that is highly expressed in brain. This kinase contains several protein-protein interaction domains and its action is partially regulated by phosphorylation. As a first step to address the neuronal functions of Ack1, here we screened mouse brain samples to identify proteins that interact with this kinase. Using mass spectrometry analysis, we identified new putative partners for Ack1 including cytoskeletal proteins such as Drebrin or MAP4; adhesion regulators such as NCAM1 and neurabin-2; and synapse mediators such as SynGAP, GRIN1 and GRIN3. In addition, we confirmed that Ack1 and CAMKII both co-immunoprecipitate and co-localize in neurons. We also identified that adult and P5 samples contained the phosphorylated residues Thr 104 and Ser 825, and only P5 samples contained phosphorylated Ser 722, a site linked to cancer and interleukin signaling when phosphorylated. All these findings support the notion that Ack1 could be involved in neuronal plasticity.
Collapse
Affiliation(s)
- Maria Del Mar Masdeu
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain.,Present address: Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London NW7 1AA, United Kingdom
| | - Beatriz G Armendáriz
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Anna La Torre
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Present address: Department of Cell Biology and Human Anatomy, University of California Davis, 95616 Davis, California, USA
| | - Eduardo Soriano
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain.,Vall d´Hebron Institute of Research, Barcelona 08035, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Ferran Burgaya
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Jesús Mariano Ureña
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| |
Collapse
|
37
|
Verduzco D, Kuenzi BM, Kinose F, Sondak VK, Eroglu Z, Rix U, Smalley KSM. Ceritinib Enhances the Efficacy of Trametinib in BRAF/NRAS-Wild-Type Melanoma Cell Lines. Mol Cancer Ther 2017; 17:73-83. [PMID: 29133622 DOI: 10.1158/1535-7163.mct-17-0196] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/06/2017] [Accepted: 10/13/2017] [Indexed: 11/16/2022]
Abstract
Targeted therapy options are currently lacking for the heterogeneous population of patients whose melanomas lack BRAF or NRAS mutations (∼35% of cases). We undertook a chemical biology screen to identify potential novel drug targets for this understudied group of tumors. Screening a panel of 8 BRAF/NRAS-WT melanoma cell lines against 240 targeted drugs identified ceritinib and trametinib as potential hits with single-agent activity. Ceritinib enhanced the efficacy of trametinib across the majority of the BRAF/NRAS-WT cell lines, and the combination showed increased cytotoxicity in both three-dimensional spheroid culture and long-term colony formation experiments. Coadministration of ceritinib and trametinib led to robust inhibition of tumor growth in an in vivo xenograft BRAF/NRAS-WT melanoma model; this was not due to ALK inhibition by ceritinib. Mechanistic studies showed the ceritinib-trametinib combination to increase suppression of MAPK and TORC1 signaling. Similar results were seen when BRAF/NRAS-WT melanoma cells were treated with a combination of trametinib and the TORC1/2 inhibitor INK128. We next used mass spectrometry-based chemical proteomics and identified known and new ceritinib targets, such as IGF1R and ACK1, respectively. Validation studies suggested that ceritinib could suppress mTORC1 signaling in the presence of trametinib through inhibition of IGF1R and/or ACK1 in a cell line-dependent manner. Together, our studies demonstrated that combining a specific inhibitor (trametinib) with a more broadly targeted agent (ceritinib) has efficacy against tumors with heterogeneous mutational profiles. Mol Cancer Ther; 17(1); 73-83. ©2017 AACR.
Collapse
Affiliation(s)
- Daniel Verduzco
- The Department of Tumor Biology, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brent M Kuenzi
- The Department of Drug Discovery, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Fumi Kinose
- The Department of Drug Discovery, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Vernon K Sondak
- The Department of Cutaneous Oncology, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Zeynep Eroglu
- The Department of Cutaneous Oncology, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Uwe Rix
- The Department of Drug Discovery, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Keiran S M Smalley
- The Department of Tumor Biology, The Moffitt Cancer Center and Research Institute, Tampa, Florida. .,The Department of Cutaneous Oncology, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
38
|
Sun A, Wei J, Childress C, Shaw JH, Peng K, Shao G, Yang W, Lin Q. The E3 ubiquitin ligase NEDD4 is an LC3-interactive protein and regulates autophagy. Autophagy 2017; 13:522-537. [PMID: 28085563 DOI: 10.1080/15548627.2016.1268301] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The MAP1LC3/LC3 family plays an essential role in autophagosomal biogenesis and transport. In this report, we show that the HECT family E3 ubiquitin ligase NEDD4 interacts with LC3 and is involved in autophagosomal biogenesis. NEDD4 binds to LC3 through a conserved WXXL LC3-binding motif in a region between the C2 and the WW2 domains. Knockdown of NEDD4 impaired starvation- or rapamycin-induced activation of autophagy and autophagosomal biogenesis and caused aggregates of the LC3 puncta colocalized with endoplasmic reticulum membrane markers. Electron microscopy observed gigantic deformed mitochondria in NEDD4 knockdown cells, suggesting that NEDD4 might function in mitophagy. Furthermore, SQSTM1 is ubiquitinated by NEDD4 while LC3 functions as an activator of NEDD4 ligase activity. Taken together, our studies define an important role of NEDD4 in regulation of autophagy.
Collapse
Affiliation(s)
- Aiqin Sun
- a School of Medicine, Jiangsu University , Zhenjiang , China
| | - Jing Wei
- a School of Medicine, Jiangsu University , Zhenjiang , China
| | - Chandra Childress
- b Department of Biology , Susquehanna University , Selinsgrove , PA , USA
| | - John H Shaw
- c Department of Pathology , Geisinger Clinic , Danville , PA , USA
| | - Ke Peng
- a School of Medicine, Jiangsu University , Zhenjiang , China
| | - Genbao Shao
- a School of Medicine, Jiangsu University , Zhenjiang , China
| | - Wannian Yang
- a School of Medicine, Jiangsu University , Zhenjiang , China
| | - Qiong Lin
- a School of Medicine, Jiangsu University , Zhenjiang , China
| |
Collapse
|
39
|
Modular transcriptional repertoire and MicroRNA target analyses characterize genomic dysregulation in the thymus of Down syndrome infants. Oncotarget 2016; 7:7497-533. [PMID: 26848775 PMCID: PMC4884935 DOI: 10.18632/oncotarget.7120] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/23/2016] [Indexed: 12/25/2022] Open
Abstract
Trisomy 21-driven transcriptional alterations in human thymus were characterized through gene coexpression network (GCN) and miRNA-target analyses. We used whole thymic tissue--obtained at heart surgery from Down syndrome (DS) and karyotipically normal subjects (CT)--and a network-based approach for GCN analysis that allows the identification of modular transcriptional repertoires (communities) and the interactions between all the system's constituents through community detection. Changes in the degree of connections observed for hierarchically important hubs/genes in CT and DS networks corresponded to community changes. Distinct communities of highly interconnected genes were topologically identified in these networks. The role of miRNAs in modulating the expression of highly connected genes in CT and DS was revealed through miRNA-target analysis. Trisomy 21 gene dysregulation in thymus may be depicted as the breakdown and altered reorganization of transcriptional modules. Leading networks acting in normal or disease states were identified. CT networks would depict the "canonical" way of thymus functioning. Conversely, DS networks represent a "non-canonical" way, i.e., thymic tissue adaptation under trisomy 21 genomic dysregulation. This adaptation is probably driven by epigenetic mechanisms acting at chromatin level and through the miRNA control of transcriptional programs involving the networks' high-hierarchy genes.
Collapse
|
40
|
Chapter Six - The Ubiquitin Network in the Control of EGFR Endocytosis and Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:225-76. [DOI: 10.1016/bs.pmbts.2016.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
41
|
Wu X, Zahari MS, Ma B, Liu R, Renuse S, Sahasrabuddhe NA, Chen L, Chaerkady R, Kim MS, Zhong J, Jelinek C, Barbhuiya MA, Leal-Rojas P, Yang Y, Kashyap MK, Marimuthu A, Ling M, Fackler MJ, Merino V, Zhang Z, Zahnow CA, Gabrielson E, Stearns V, Roa JC, Sukumar S, Gill PS, Pandey A. Global phosphotyrosine survey in triple-negative breast cancer reveals activation of multiple tyrosine kinase signaling pathways. Oncotarget 2015; 6:29143-60. [PMID: 26356563 PMCID: PMC4745717 DOI: 10.18632/oncotarget.5020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/24/2015] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most prevalent cancer in women worldwide. About 15-20% of all breast cancers are triple negative breast cancer (TNBC) and are often highly aggressive when compared to other subtypes of breast cancers. To better characterize the biology that underlies the TNBC phenotype, we profiled the phosphotyrosine proteome of a panel of twenty-six TNBC cell lines using quantitative high resolution Fourier transform mass spectrometry. A heterogeneous pattern of tyrosine kinase activation was observed based on 1,789 tyrosine-phosphorylated peptides identified from 969 proteins. One of the tyrosine kinases, AXL, was found to be activated in a majority of aggressive TNBC cell lines and was accompanied by a higher level of AXL expression. High levels of AXL expression are correlated with a significant decrease in patient survival. Treatment of cells bearing activated AXL with a humanized AXL antibody inhibited cell proliferation and migration in vitro, and tumor growth in mice. Overall, our global phosphoproteomic analysis provided new insights into the heterogeneity in the activation status of tyrosine kinase pathways in TNBCs. Our approach presents an effective means of identifying important novel biomarkers and targets for therapy such as AXL in TNBC.
Collapse
Affiliation(s)
- Xinyan Wu
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Muhammad Saddiq Zahari
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Binyun Ma
- 6 Department of Medicine, University of Southern California, Los Angeles, USA
| | - Ren Liu
- 6 Department of Medicine, University of Southern California, Los Angeles, USA
| | - Santosh Renuse
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 5 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Nandini A. Sahasrabuddhe
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 5 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Lily Chen
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Raghothama Chaerkady
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Min-Sik Kim
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jun Zhong
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Christine Jelinek
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Mustafa A. Barbhuiya
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 5 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Pamela Leal-Rojas
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 7 Department of Pathology, Center of Genetic and Immunological Studies (CEGIN) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Yi Yang
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Manoj Kumar Kashyap
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 5 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Arivusudar Marimuthu
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 5 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Min Ling
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Mary Jo Fackler
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Vanessa Merino
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Zhen Zhang
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Cynthia A. Zahnow
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Edward Gabrielson
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
- 4 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Vered Stearns
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Juan Carlos Roa
- 8 Advanced Center for Chronic Diseases (ACCDiS), Department of Pathology Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Saraswati Sukumar
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Parkash S. Gill
- 6 Department of Medicine, University of Southern California, Los Angeles, USA
| | - Akhilesh Pandey
- 1 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, USA
- 2 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- 3 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
- 4 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
42
|
Park E, Kim N, Ficarro SB, Zhang Y, Lee BI, Cho A, Kim K, Park AK, Park WY, Murray B, Meyerson M, Beroukhim R, Marto JA, Cho J, Eck MJ. Structure and mechanism of activity-based inhibition of the EGF receptor by Mig6. Nat Struct Mol Biol 2015; 22:703-711. [PMID: 26280531 PMCID: PMC4790445 DOI: 10.1038/nsmb.3074] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022]
Abstract
Mig6 is a feedback inhibitor that directly binds, inhibits and drives internalization of ErbB-family receptors. Mig6 selectively targets activated receptors. Here we found that the epidermal growth factor receptor (EGFR) phosphorylates Mig6 on Y394 and that this phosphorylation is primed by prior phosphorylation of an adjacent residue, Y395, by Src. Crystal structures of human EGFR-Mig6 complexes reveal the structural basis for enhanced phosphorylation of primed Mig6 and show how Mig6 rearranges after phosphorylation by EGFR to effectively irreversibly inhibit the same receptor that catalyzed its phosphorylation. This dual phosphorylation site allows Mig6 to inactivate EGFR in a manner that requires activation of the target receptor and that can be modulated by Src. Loss of Mig6 is a driving event in human cancer; analysis of 1,057 gliomas reveals frequent focal deletions of ERRFI1, the gene that encodes Mig6, in EGFR-amplified glioblastomas.
Collapse
Affiliation(s)
- Eunyoung Park
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA USA
| | - Nayoung Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, Republic of Korea
| | - Scott B. Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA USA
| | - Yi Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA USA
| | - Byung Il Lee
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Biomolecular Function Research Branch, Division of Convergence Technology, Research Institute, National Cancer Center, Goyang, Gyeonggi Republic of Korea
| | - Ahye Cho
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, Republic of Korea
| | - Kihong Kim
- Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, Republic of Korea
| | - Angela K.J. Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, Republic of Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, Republic of Korea
| | | | - Matthew Meyerson
- Broad Institute of Harvard and MIT, Cambridge, MA USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Pathology, Harvard Medical School, Boston, MA USA
| | - Rameen Beroukhim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Broad Institute of Harvard and MIT, Cambridge, MA USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Medicine, Harvard Medical School, Boston, MA USA
| | - Jarrod A. Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA USA
| | - Jeonghee Cho
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, SungKyunKwan University, Seoul, Republic of Korea
| | - Michael J. Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA USA
| |
Collapse
|
43
|
Hu F, Liu H, Xie X, Mei J, Wang M. Activated cdc42-associated kinase is up-regulated in non-small-cell lung cancer and necessary for FGFR-mediated AKT activation. Mol Carcinog 2015; 55:853-63. [PMID: 25945695 DOI: 10.1002/mc.22327] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 12/09/2014] [Accepted: 03/26/2015] [Indexed: 11/08/2022]
Abstract
Activated cdc42-associated tyrosine kinase 1 (ACK1) has been reported to be implicated in non-small-cell lung cancer (NSCLC). However, the expression pattern and biological functions of ACK1 in the progression of NSCLC are not fully understood. In this study, it was found that the expression of ACK1 was significantly up-regulated in NSCLC samples compared to their adjacent normal tissues. Meanwhile, the expression of ACK1 was inversely correlated with the survival of NSCLC patients. Moreover, in the biological function studies, ACK1 was further validated to promote the growth, migration, and metastasis of NSCLC cells in vitro and in vivo. Mechanistically, ACK1 bind with FGFR1 and was essential for the phosphorylation of AKT induced by FGF. Our study demonstrated that ACK1 played an oncogenic role in the progression of NSCLC and ACK1 might be a promising target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Fengqing Hu
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hongcheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao Xie
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Ju Mei
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Mingsong Wang
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Affiliation(s)
- Muhammad Imran Qadir
- Institute of Molecular Biology and Biotechnology; Bahauddin Zakariya University; Multan Pakistan
| | - Amna Parveen
- Pharmaceutical Resources Botany Lab; Department of Pharmacognosy; College of Pharmacy; Chung-Ang University; Seoul South Korea
| | - Muhammad Ali
- Government College University Faisalabad; Faisalabad Pakistan
| |
Collapse
|
45
|
Chung BM, Tom E, Zutshi N, Bielecki TA, Band V, Band H. Nexus of signaling and endocytosis in oncogenesis driven by non-small cell lung cancer-associated epidermal growth factor receptor mutants. World J Clin Oncol 2014; 5:806-823. [PMID: 25493220 PMCID: PMC4259944 DOI: 10.5306/wjco.v5.i5.806] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/19/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) controls a wide range of cellular processes, and aberrant EGFR signaling as a result of receptor overexpression and/or mutation occurs in many types of cancer. Tumor cells in non-small cell lung cancer (NSCLC) patients that harbor EGFR kinase domain mutations exhibit oncogene addiction to mutant EGFR, which confers high sensitivity to tyrosine kinase inhibitors (TKIs). As patients invariably develop resistance to TKIs, it is important to delineate the cell biological basis of mutant EGFR-induced cellular transformation since components of these pathways can serve as alternate therapeutic targets to preempt or overcome resistance. NSCLC-associated EGFR mutants are constitutively-active and induce ligand-independent transformation in nonmalignant cell lines. Emerging data suggest that a number of factors are critical for the mutant EGFR-dependent tumorigenicity, and bypassing the effects of TKIs on these pathways promotes drug resistance. For example, activation of downstream pathways such as Akt, Erk, STAT3 and Src is critical for mutant EGFR-mediated biological processes. It is now well-established that the potency and spatiotemporal features of cellular signaling by receptor tyrosine kinases such as EGFR, as well as the specific pathways activated, is determined by the nature of endocytic traffic pathways through which the active receptors traverse. Recent evidence indicates that NSCLC-associated mutant EGFRs exhibit altered endocytic trafficking and they exhibit reduced Cbl ubiquitin ligase-mediated lysosomal downregulation. More recent work has shown that mutant EGFRs undergo ligand-independent traffic into the endocytic recycling compartment, a behavior that plays a key role in Src pathway activation and oncogenesis. These studies are beginning to delineate the close nexus between signaling and endocytic traffic of EGFR mutants as a key driver of oncogenic processes. Therefore, in this review, we will discuss the links between mutant EGFR signaling and endocytic properties, and introduce potential mechanisms by which altered endocytic properties of mutant EGFRs may alter signaling and vice versa as well as their implications for NSCLC therapy.
Collapse
|
46
|
Amarachintha SP, Ryan KJ, Cayer M, Boudreau NS, Johnson NM, Heckman CA. Effect of Cdc42 domains on filopodia sensing, cell orientation, and haptotaxis. Cell Signal 2014; 27:683-93. [PMID: 25435426 DOI: 10.1016/j.cellsig.2014.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/21/2014] [Indexed: 11/17/2022]
Abstract
Filopodia are sensors which, along with microtubules, regulate the persistence of locomotion. To determine whether protrusions were involved in sensing adhesion, epithelial cells were cultured on platinum and tantalum gradients. Protrusions were defined by an unbiased statistical method of classification as factors 4 (filopodia), 5 (mass distribution), and 7 (nascent neurites). When the prevalence of protrusions was measured in zones of high (H), middle (M), and low (L) adhesiveness, the main differences were in factor 4. Its values were highest at H and declined at M and L regardless of the gradient composition. The significance of the differences was enhanced when T (top/adhesive end) and B (bottom/nonadhesive end) sides of cells were analyzed separately. Since information about sidedness increased the statistical power of the test, this result suggested that cells pointed more filopodia toward the adhesive end. Trends occurred in factors 5 and 7 only when conditions allowed for a marked trend in factor 4. The data showed that gradient sensing is proportional to the prevalence of filopodia, and filopodia are the only protrusions engaged in comparing adhesiveness across a cell. The probability (P) of the significance of a trend was then used to determine how cells sense the gradient. Binding peptides (BPs) were introduced representing sequences critical for Cdc42 docking on a specific partner. BPs for IQGAP (IQ(calmodulin-binding domain)-containing GTPase-activating protein) and ACK (Cdc42-associated kinase) reduced factor 4 values and prevented cell orientation on the gradient. Micrographs showed attenuated or stubby filopodia. These effectors may be implicated in gradient sensing. Another IQGAP BP increased filopodia prevalence and enhanced orientation on the gradient (P<0.00015). A Wiskott-Aldrich syndrome protein (WASP) BP had no effect. When sensing and orientation were abolished, they both failed at the level of filopodia, indicating that filopodia are both sensors and implementers of signals transduced by adhesion.
Collapse
Affiliation(s)
- Surya P Amarachintha
- Center for Microscopy and Microanalysis, Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Kenneth J Ryan
- Department of Applied Statistics and Operations Research, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Marilyn Cayer
- Center for Microscopy and Microanalysis, Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Nancy S Boudreau
- Department of Applied Statistics and Operations Research, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Nathan M Johnson
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Carol A Heckman
- Center for Microscopy and Microanalysis, Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, United States; Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, United States
| |
Collapse
|
47
|
Rudd ML, Mohamed H, Price JC, O'Hara AJ, Le Gallo M, Urick ME, Cruz P, Zhang S, Hansen NF, Godwin AK, Sgroi DC, Wolfsberg TG, Mullikin JC, Merino MJ, Bell DW. Mutational analysis of the tyrosine kinome in serous and clear cell endometrial cancer uncovers rare somatic mutations in TNK2 and DDR1. BMC Cancer 2014; 14:884. [PMID: 25427824 PMCID: PMC4258955 DOI: 10.1186/1471-2407-14-884] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 11/13/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endometrial cancer (EC) is the 8th leading cause of cancer death amongst American women. Most ECs are endometrioid, serous, or clear cell carcinomas, or an admixture of histologies. Serous and clear ECs are clinically aggressive tumors for which alternative therapeutic approaches are needed. The purpose of this study was to search for somatic mutations in the tyrosine kinome of serous and clear cell ECs, because mutated kinases can point to potential therapeutic targets. METHODS In a mutation discovery screen, we PCR amplified and Sanger sequenced the exons encoding the catalytic domains of 86 tyrosine kinases from 24 serous, 11 clear cell, and 5 mixed histology ECs. For somatically mutated genes, we next sequenced the remaining coding exons from the 40 discovery screen tumors and sequenced all coding exons from another 72 ECs (10 clear cell, 21 serous, 41 endometrioid). We assessed the copy number of mutated kinases in this cohort of 112 tumors using quantitative real time PCR, and we used immunoblotting to measure expression of these kinases in endometrial cancer cell lines. RESULTS Overall, we identified somatic mutations in TNK2 (tyrosine kinase non-receptor, 2) and DDR1 (discoidin domain receptor tyrosine kinase 1) in 5.3% (6 of 112) and 2.7% (3 of 112) of ECs. Copy number gains of TNK2 and DDR1 were identified in another 4.5% and 0.9% of 112 cases respectively. Immunoblotting confirmed TNK2 and DDR1 expression in endometrial cancer cell lines. Three of five missense mutations in TNK2 and one of two missense mutations in DDR1 are predicted to impact protein function by two or more in silico algorithms. The TNK2(P761Rfs*72) frameshift mutation was recurrent in EC, and the DDR1(R570Q) missense mutation was recurrent across tumor types. CONCLUSIONS This is the first study to systematically search for mutations in the tyrosine kinome in clear cell endometrial tumors. Our findings indicate that high-frequency somatic mutations in the catalytic domains of the tyrosine kinome are rare in clear cell ECs. We uncovered ten new mutations in TNK2 and DDR1 within serous and endometrioid ECs, thus providing novel insights into the mutation spectrum of each gene in EC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Daphne W Bell
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda MD 20892, USA.
| |
Collapse
|
48
|
Mahajan K, Mahajan NP. ACK1/TNK2 tyrosine kinase: molecular signaling and evolving role in cancers. Oncogene 2014; 34:4162-7. [PMID: 25347744 PMCID: PMC4411206 DOI: 10.1038/onc.2014.350] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 01/11/2023]
Abstract
Deregulated tyrosine kinase signaling alters cellular homeostasis to drive cancer progression. The emergence of a non-receptor tyrosine kinase, ACK1 as an oncogenic kinase, has uncovered novel mechanisms by which tyrosine kinase signaling promotes cancer progression. While early studies focused on ACK1 (also known as activated Cdc42-associated kinase 1 or TNK2) as a cytosolic effecter of activated transmembrane receptor tyrosine kinases (RTKs), wherein it shuttles between the cytosol and the nucleus to rapidly transduce extracellular signals from the RTKs to the intracellular effectors, recent data unfold a new aspect of its functionality as an epigenetic regulator. ACK1 interacts with the Estrogen Receptor (ER)/histone demethylase KDM3A (JHDM2a) complex, modifies KDM3A by tyrosine phosphorylation to regulate transcriptional outcome at HOXA1 locus to promote the growth of tamoxifen-resistant breast cancer. It is also well established that ACK1 regulates the activity of Androgen Receptor (AR) by tyrosine phosphorylation to fuel the growth of hormone-refractory prostate cancers. Further, recent explosion in genomic sequencing has revealed recurrent ACK1 gene amplification and somatic mutations in a variety of human malignancies, providing a molecular basis for its role in neoplastic transformation. In this review, we will discuss the various facets of ACK1 signaling, including its newly uncovered epigenetic regulator function, which enables cells to bypass the blockade to major survival pathways to promote resistance to standard cancer treatments. Not surprisingly, cancer cells appear to acquire an `addiction’ to ACK1 mediated survival, particularly under stress conditions, such as growth factor deprivation or genotoxic insults or hormone deprivation. With the accelerated development of potent and selective ACK1 inhibitors, targeted treatment for cancers harboring aberrant ACK1 activity may soon become a clinical reality.
Collapse
Affiliation(s)
- K Mahajan
- 1] Moffitt Cancer Center, Drug Discovery Department, Tampa, FL, USA [2] Department of Oncologic Sciences, University of South Florida, Tampa, FL, USA
| | - N P Mahajan
- 1] Moffitt Cancer Center, Drug Discovery Department, Tampa, FL, USA [2] Department of Oncologic Sciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
49
|
Mi W, Lin Q, Childress C, Sudol M, Robishaw J, Berlot CH, Shabahang M, Yang W. Geranylgeranylation signals to the Hippo pathway for breast cancer cell proliferation and migration. Oncogene 2014; 34:3095-106. [PMID: 25109332 DOI: 10.1038/onc.2014.251] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 06/09/2014] [Accepted: 06/15/2014] [Indexed: 12/17/2022]
Abstract
Protein geranylgeranylation (GGylation) is an important biochemical process for many cellular signaling molecules. Previous studies have shown that GGylation is essential for cell survival in many types of cancer. However, the molecular mechanism mediating the cell survival effect remains elusive. In this report, we show that the Hippo pathway mediates GGylation-dependent cell proliferation and migration in breast cancer cells. Blockade of GGylation enhanced phosphorylation of Mst1/2 and Lats1, and inhibited YAP and TAZ activity and the Hippo-YAP/TAZ pathway-dependent transcription. The effect of GGylation blockade on inhibition of breast cancer cell proliferation and migration is dependent on the Hippo-YAP/TAZ signaling, in which YAP appears to regulate cell proliferation and TAZ to regulate cell migration. Furthermore, GGylation-dependent cell proliferation is correlated with the activity of YAP/TAZ in breast cancer cells. Finally, Gγ and RhoA are the GGylated proteins that may transduce GGylation signals to the Hippo-YAP/TAZ pathway. Taken together, our studies have demonstrated that the Hippo-YAP/TAZ pathway is essential for GGylation-dependent cancer cell proliferation and migration.
Collapse
Affiliation(s)
- W Mi
- Weis Center for Research, Danville, PA, USA
| | - Q Lin
- 1] Weis Center for Research, Danville, PA, USA [2] School of Medical Sciences and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | | | - M Sudol
- 1] Weis Center for Research, Danville, PA, USA [2] Department of Medicine, Mount Sinai Medical School, New York, NY, USA
| | - J Robishaw
- Weis Center for Research, Danville, PA, USA
| | - C H Berlot
- Weis Center for Research, Danville, PA, USA
| | - M Shabahang
- Department of General Surgery, Geisinger Clinic, Danville, PA, USA
| | - W Yang
- Weis Center for Research, Danville, PA, USA
| |
Collapse
|
50
|
Bosutinib inhibits migration and invasion via ACK1 in KRAS mutant non-small cell lung cancer. Mol Cancer 2014; 13:13. [PMID: 24461128 PMCID: PMC3930897 DOI: 10.1186/1476-4598-13-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/21/2014] [Indexed: 01/13/2023] Open
Abstract
The advent of effective targeted therapeutics has led to increasing emphasis on precise biomarkers for accurate patient stratification. Here, we describe the role of ACK1, a non-receptor tyrosine kinase in abrogating migration and invasion in KRAS mutant lung adenocarcinoma. Bosutinib, which inhibits ACK1 at 2.7 nM IC50, was found to inhibit cell migration and invasion but not viability in a panel of non-small cell lung cancer (NSCLC) cell lines. Knockdown of ACK1 abrogated bosutinib-induced inhibition of cell migration and invasion specifically in KRAS mutant cells. This finding was further confirmed in an in vivo zebrafish metastatic model. Tissue microarray data on 210 Singaporean lung adenocarcinomas indicate that cytoplasmic ACK1 was significantly over-expressed relative to paired adjacent non-tumor tissue. Interestingly, ACK1 expression in “normal” tissue adjacent to tumour, but not tumour, was independently associated with poor overall and relapse-free survival. In conclusion, inhibition of ACK1 with bosutinib attenuates migration and invasion in the context of KRAS mutant NSCLC and may fulfil a therapeutic niche through combinatorial treatment approaches.
Collapse
|