1
|
Li R, Yan X, Zhong W, Zheng J, Li X, Liang J, Hu Z, Liu H, Chen G, Yang Y, Zhang J, Qu E, Liu W. Stratifin promotes the malignant progression of HCC via binding and hyperactivating AKT signaling. Cancer Lett 2024; 592:216761. [PMID: 38490326 DOI: 10.1016/j.canlet.2024.216761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/06/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignant tumor with limited treatment options and poor prognosis. In this study, we reveal the pivotal role of Stratifin (SFN), also recognized as 14-3-3σ, in driving HCC progression. Our investigation underscores a substantial upregulation of SFN within HCC tissues, manifesting a significant association with worse prognostic outcomes among HCC patients. In vitro and in vivo experiments reveal that SFN overexpression significantly amplifies proliferation, mitigates sorafenib-induced effects on HCC cells, and enhances tumorigenesis. While SFN silencing exerts converse effects on HCC progression. Additionally, we unveil a critical interaction between SFN and AKT, where SFN boosts AKT kinase activity by disrupting the binding of PHLPP2 and AKT, thereby intensifying the malignant progression of HCC cells. In conclusion, this study identifies the oncogenic role of SFN and elucidates the regulatory mechanism of the SFN/AKT axis in HCC, which may provide valuable insights into the mechanisms of HCC progression and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Rong Li
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China; Guangdong Province Engineering Laboratory for Transplantation Medicine, Organ Transplantation Research Center of Guangdong Province, Guangzhou, 510630, China
| | - Xijing Yan
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Wenhui Zhong
- Department of Pancreatic and Gastric Surgery, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xuejiao Li
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Jinliang Liang
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Zhongying Hu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Huanyi Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Guihua Chen
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China; Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yang Yang
- Guangdong Province Engineering Laboratory for Transplantation Medicine, Organ Transplantation Research Center of Guangdong Province, Guangzhou, 510630, China; Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Jianwei Zhang
- Department of Pancreatic and Gastric Surgery, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Enze Qu
- Department of Ultrasound, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Wei Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China; Guangdong Province Engineering Laboratory for Transplantation Medicine, Organ Transplantation Research Center of Guangdong Province, Guangzhou, 510630, China.
| |
Collapse
|
2
|
Ge G, Wen Y, Li P, Guo Z, Liu Z. Single-Cell Plasmonic Immunosandwich Assay Reveals the Modulation of Nucleocytoplasmic Localization Fluctuation of ABL1 on Cell Migration. Anal Chem 2023; 95:17502-17512. [PMID: 38050674 DOI: 10.1021/acs.analchem.3c02593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Cell migration is an essential process of cancer metastasis. The spatiotemporal dynamics of signaling molecules influences cellular phenotypic outcomes. It has been increasingly documented that the Abelson (ABL) family kinases play critical roles in solid tumors. However, ABL1's shuttling dynamics in cell migration still remains unexplored. This is mainly because tools permitting the investigation of translocation dynamics of proteins in single living cells are lacking. Herein, to bridge this gap, we developed a unique multifunctional integrated single-cell analysis method that enables long-term observation of cell migration behavior and monitoring of signaling proteins and complexes at the subcellular level. We found that the shuttling of ABL1's to the cytoplasm results in a higher migration speed, while its trafficking back to the nucleus leads to a lower one. Furthermore, our results indicated that fluctuant protein-protein interactions between 14-3-3 and ABL1 modulate ABL1's nucleocytoplasmic fluctuation and eventually affect the cell speed. Importantly, based on these new insights, we demonstrated that disturbing ABL1's nuclear export traffic and 14-3-3-ABL1 complexes formation can effectively suppress cell migration. Thus, our method opens up a new possibility for simultaneous tracking of internal molecular mechanisms and cell behavior, providing a promising tool for the in-depth study of cancer.
Collapse
Affiliation(s)
- Ge Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Yanrong Wen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Pengfei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhanchen Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| |
Collapse
|
3
|
Kor A, Yalçın M, Erten Ş, Maraş Y, Oğuz EF, Doğan İ, Atalar E, Başer S, Erel Ö. 14-3-3η Proteins as a Diagnostic Marker, Disease Activation Indicator, and Lymphoma Predictor in Patients with Primary Sjögren Syndrome. ARCHIVES OF IRANIAN MEDICINE 2023; 26:582-591. [PMID: 38310415 PMCID: PMC10862092 DOI: 10.34172/aim.2023.85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 07/03/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Primary Sjögren syndrome (PSS) is a chronic, autoimmune, and lymphoproliferative disease of the connective tissue. In patients with PSS, the risk of developing B-cell non-Hodgkin lymphoma (NHL) increases dramatically, with a prevalence of approximately 5%. The 14-3-3 protein isoforms are phospho-serin/phospho-threonine binding proteins associated with many malignant diseases. This study aimed to evaluate the relationship between disease activity parameters and markers predicting lymphoma development in patients with PSS and 14-3-3η proteins. METHODS This study was designed as an analytical case-control study. A total of 57 PSS patients and 54 healthy volunteers were included in the study. The European League Against Rheumatism (EULAR) Sjögren syndrome disease activity index (ESSDAI) was used to assess systemic disease activity in PSS. Receiver operating characteristic (ROC) analysis was used to test the diagnostic accuracy measures of the analytical results. Multivariable linear regression analysis was used to evaluate the effects of independent variables on the 14-3-3η protein. RESULTS The 14-3-3η protein serum levels were found to be significantly higher in PSS (2.72 [2.04-4.07]) than healthy controls (1.73 [1.41-2.43]) (P<0.0001). A significant relationship was found between 14-3-3η protein levels and ESSDAI group (β=0.385, 95%CI=0.318-1.651, P=0.005), hypocomplementemia (C3 or C4) (β=0.223, 95% CI=0.09-1.983, P=0.048) and purpura (β=0.252, 95% CI=0.335-4.903, P=0.022), which are accepted as lymphoma predictors. A significant correlation was found between PSS disease activity score ESSDAI and 14-33η protein (β=0.496, 95% CI=0.079-0.244, P=0.0002). CONCLUSION 14-3-3η proteins are potential candidates for diagnostic marker, marker of disease activity, and predictor of lymphoma in PSS patients.
Collapse
Affiliation(s)
- Ahmet Kor
- Department of Rheumatology, Aksaray Education and Research Hospital, Aksaray, Turkey
| | - Merve Yalçın
- Department of Internal Medicine, Ankara Bilkent City Hospital, Ministry of Health, Ankara, Turkey
| | - Şükran Erten
- Department of Rheumatology, Faculty of Medicine Ankara Bilkent City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Yüksel Maraş
- Department of Rheumatology, Ankara Bilkent City Hospital, Health Sciences University, Ankara, Turkey
| | - Esra Fırat Oğuz
- Department of Medical Biochemistry, Ankara Bilkent City Hospital, Ministry of Health, Ankara, Turkey
| | - İsmail Doğan
- Department of Rheumatology, Faculty of Medicine Ankara Bilkent City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Ebru Atalar
- Department of Rheumatology, Ankara Bilkent City Hospital, Ministry of Health, Ankara, Turkey
| | - Salih Başer
- Department of Internal Medicine, Faculty of Medicine Ankara Bilkent City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Özcan Erel
- Department of Medical Biochemistry, Faculty of Medicine Ankara Bilkent City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
4
|
Deb Roy A, Gross EG, Pillai GS, Seetharaman S, Etienne-Manneville S, Inoue T. Non-catalytic allostery in α-TAT1 by a phospho-switch drives dynamic microtubule acetylation. J Cell Biol 2022; 221:213540. [PMID: 36222836 PMCID: PMC9565784 DOI: 10.1083/jcb.202202100] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/03/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Spatiotemporally dynamic microtubule acetylation underlies diverse physiological and pathological events. Despite its ubiquity, the molecular mechanisms that regulate the sole microtubule acetylating agent, α-tubulin-N-acetyltransferase-1 (α-TAT1), remain obscure. Here, we report that dynamic intracellular localization of α-TAT1 along with its catalytic activity determines efficiency of microtubule acetylation. Specifically, we newly identified a conserved signal motif in the intrinsically disordered C-terminus of α-TAT1, consisting of three competing regulatory elements-nuclear export, nuclear import, and cytosolic retention. Their balance is tuned via phosphorylation by CDK1, PKA, and CK2, and dephosphorylation by PP2A. While the unphosphorylated form binds to importins and resides both in cytosol and nucleus, the phosphorylated form binds to specific 14-3-3 adapters and accumulates in the cytosol for maximal substrate access. Unlike other molecules with a similar phospho-regulated signal motif, α-TAT1 uniquely uses the nucleus as a hideout. This allosteric spatial regulation of α-TAT1 function may help uncover a spatiotemporal code of microtubule acetylation in normal and aberrant cell behavior.
Collapse
Affiliation(s)
- Abhijit Deb Roy
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691, Université Paris Cité, Centre national de la recherche scientifique, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691, Université Paris Cité, Centre national de la recherche scientifique, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Takanari Inoue
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
5
|
Effect of HSP90AB1 and CC domain interaction on Bcr-Abl protein cytoplasm localization and function in chronic myeloid leukemia cells. Cell Commun Signal 2021; 19:71. [PMID: 34217296 PMCID: PMC8254927 DOI: 10.1186/s12964-021-00752-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 05/21/2021] [Indexed: 11/10/2022] Open
Abstract
Background The fusion oncoprotein Bcr-Abl is mostly located in the cytoplasm, which causes chronic myeloid leukemia (CML). After moving into the nucleus, the fusion protein can induce apoptosis of CML cells. The coiled-coil domain (CC domain) of Bcr-Abl protein plays a central role in the subcellular localization. However, how CC domain affects subcellular localization of Bcr-Abl remains unclear. Methods Herein, the key proteins interacting with the Bcr-Abl CC domain were screened by immunoprecipitation binding mass spectrometry. The specific site of Bcr-Abl CC domain binding to target protein was predicted by Deep Viewer. Immunoprecipitation assay was used to confirmed the specific sites of protein binding. IF and western blot were used to observe the subcellular localization of target protein. Western blot was used to examine the protein changes. CCK-8, clonal formation test and FCM cycle detection were used to observe the effect of inhibitor on the proliferation ability of CML cells. FCM apoptosis detection was used to observe the level of cells apoptosis. Results HSP90AB1 interacts with Bcr-Abl CC domain via N-terminal domain (NTD), preventing the transport of Bcr-Abl protein to the nucleus and maintaining the activation of Bcr-Abl tyrosine kinase. The nucleus-entrapped Bcr-Abl markedly inhibits the proliferation and induces apoptosis of CML cells by activating p73 and repressing the expression of cytoplasmic oncogenic signaling pathways mediated by Bcr-Abl. Moreover, the combination of 17AAG (Tanespimycin) with Leptomycin B (LMB) considerably decreased the proliferation of CML cells. Conclusion Our study provides evidence that it is feasible to transport Bcr-Abl into the nucleus as an alternative strategy for the treatment of CML, and targeting the NTD of HSP90AB1 to inhibit the interaction with Bcr-Abl is more accurate for the development and application of HSP90 inhibitor in the treatment of CML and other Bcr-Abl-addicted malignancies. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00752-9.
Collapse
|
6
|
Aljabal G, Yap BK. 14-3-3σ and Its Modulators in Cancer. Pharmaceuticals (Basel) 2020; 13:ph13120441. [PMID: 33287252 PMCID: PMC7761676 DOI: 10.3390/ph13120441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/19/2023] Open
Abstract
14-3-3σ is an acidic homodimer protein with more than one hundred different protein partners associated with oncogenic signaling and cell cycle regulation. This review aims to highlight the crucial role of 14-3-3σ in controlling tumor growth and apoptosis and provide a detailed discussion on the structure-activity relationship and binding interactions of the most recent 14-3-3σ protein-protein interaction (PPI) modulators reported to date, which has not been reviewed previously. This includes the new fusicoccanes stabilizers (FC-NAc, DP-005), fragment stabilizers (TCF521-123, TCF521-129, AZ-003, AZ-008), phosphate-based inhibitors (IMP, PLP), peptide inhibitors (2a-d), as well as inhibitors from natural sources (85531185, 95911592). Additionally, this review will also include the discussions of the recent efforts by a different group of researchers for understanding the binding mechanisms of existing 14-3-3σ PPI modulators. The strategies and state-of-the-art techniques applied by various group of researchers in the discovery of a different chemical class of 14-3-3σ modulators for cancer are also briefly discussed in this review, which can be used as a guide in the development of new 14-3-3σ modulators in the near future.
Collapse
|
7
|
Iralde-Lorente L, Tassone G, Clementi L, Franci L, Munier CC, Cau Y, Mori M, Chiariello M, Angelucci A, Perry MWD, Pozzi C, Mangani S, Botta M. Identification of Phosphate-Containing Compounds as New Inhibitors of 14-3-3/c-Abl Protein-Protein Interaction. ACS Chem Biol 2020; 15:1026-1035. [PMID: 32142251 DOI: 10.1021/acschembio.0c00039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The 14-3-3/c-Abl protein-protein interaction (PPI) is related to carcinogenesis and in particular to pathogenesis of chronic myeloid leukemia (CML). Previous studies have demonstrated that molecules able to disrupt this interaction improve the nuclear translocation of c-Abl, inducing apoptosis in leukemia cells. Through an X-ray crystallography screening program, we have identified two phosphate-containing compounds, inosine monophosphate (IMP) and pyridoxal phosphate (PLP), as binders of human 14-3-3σ, by targeting the protein amphipathic groove. Interestingly, they also act as weak inhibitors of the 14-3-3/c-Abl PPI, demonstrated by NMR, SPR, and FP data. A 37-compound library of PLP and IMP analogues was investigated using a FP assay, leading to the identification of three further molecules acting as weak inhibitors of the 14-3-3/c-Abl complex formation. The antiproliferative activity of IMP, PLP, and the three derivatives was tested against K-562 cells, showing that the parent compounds had the most pronounced effect on tumor cells. PLP and IMP were also effective in promoting the c-Abl nuclear translocation in c-Abl overexpressing cells. Further, these compounds demonstrated low cytotoxicity on human Hs27 fibroblasts. In conclusion, our data suggest that 14-3-3σ targeting compounds represent promising hits for further development of drugs against c-Abl-dependent cancers.
Collapse
Affiliation(s)
- Leire Iralde-Lorente
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Letizia Clementi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100, L’Aquila, Italy
| | - Lorenzo Franci
- Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina, 1 53100 Siena, Italy
- Dipartimento di Biotecnologie Mediche − Dipartimento di Eccellenza 2018-2022, Università degli Studi di Siena, via Aldo Moro, 2 53100 Siena, Italy
- Consiglio Nazionale delle Ricerche, Istituto di Fisiologia Clinica, Via Fiorentina 1, 53100 Siena, Italy
| | - Claire C Munier
- Medicinal Chemistry, Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Ylenia Cau
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Mario Chiariello
- Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Via Fiorentina, 1 53100 Siena, Italy
- Consiglio Nazionale delle Ricerche, Istituto di Fisiologia Clinica, Via Fiorentina 1, 53100 Siena, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100, L’Aquila, Italy
| | - Matthew W. D. Perry
- Medicinal Chemistry, Research and Early Development, Respiratory, Inflammation and Autoimmune (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy−Department of Excellence 2018-2022, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
8
|
Chichirau BE, Diechler S, Posselt G, Wessler S. Tyrosine Kinases in Helicobacter pylori Infections and Gastric Cancer. Toxins (Basel) 2019; 11:toxins11100591. [PMID: 31614680 PMCID: PMC6832112 DOI: 10.3390/toxins11100591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori (H. pylori) has been identified as a leading cause of gastric cancer, which is one of the most frequent and malignant types of tumor. It is characterized by its rapid progression, distant metastases, and resistance to conventional chemotherapy. A number of receptor tyrosine kinases and non-receptor tyrosine kinases have been implicated in H. pylori-mediated pathogenesis and tumorigenesis. In this review, recent findings of deregulated EGFR, c-Met, JAK, FAK, Src, and c-Abl and their functions in H. pylori pathogenesis are summarized.
Collapse
Affiliation(s)
- Bianca E Chichirau
- Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| | - Sebastian Diechler
- Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| | - Gernot Posselt
- Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| | - Silja Wessler
- Cancer Cluster Salzburg, Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
9
|
Crosstalk between 14-3-3θ and AF4 enhances MLL-AF4 activity and promotes leukemia cell proliferation. Cell Oncol (Dordr) 2019; 42:829-845. [PMID: 31493143 DOI: 10.1007/s13402-019-00468-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2019] [Indexed: 01/14/2023] Open
Abstract
PURPOSE The t(4;11)(q21;q23) translocation characterizes a form of acute lymphoblastic leukemia with a poor prognosis. It results in a fusion gene encoding a chimeric transcription factor, MLL-AF4, that deregulates gene expression through a variety of still controversial mechanisms. To provide new insights into these mechanisms, we examined the interaction between AF4, the most common MLL fusion partner, and the scaffold protein 14-3-3θ, in the context of t(4;11)-positive leukemia. METHODS Protein-protein interactions were analyzed using immunoprecipitation and in vitro binding assays, and by fluorescence microscopy in t(4;11)-positive RS4;11 and MV4-11 leukemia cells and in HEK293 cells. Protein and mRNA expression levels were determined by Western blotting and RT-qPCR, respectively. A 5-bromo-2'-deoxyuridine assay and an annexin V/propidium iodide assay were used to assess proliferation and apoptosis rates, respectively, in t(4;11)-positive and control cells. Chromatin immunoprecipitation was performed to assess binding of 14-3-3θ and AF4 to a specific promoter element. RESULTS We found that AF4 and 14-3-3θ are nuclear interactors, that 14-3-3θ binds Ser588 of AF4 and that 14-3-3θ forms a complex with MLL-AF4. In addition, we found that in t(4;11)-positive cells, 14-3-3θ knockdown decreased the expression of MLL-AF4 target genes, induced apoptosis and hampered cell proliferation. Moreover, we found that 14-3-3θ knockdown impaired the recruitment of AF4, but not of MLL-AF4, to target chromatin. Overall, our data indicate that the activity of the chimeric transcription factor MLL-AF4 depends on the cellular availability of 14-3-3θ, which triggers the transactivating function and subsequent degradation of AF4. CONCLUSIONS From our data we conclude that the scaffold protein 14-3-3θ enhances the aberrant activity of the chimeric transcription factor MLL-AF4 and, therefore, represents a new player in the molecular pathogenesis of t(4;11)-positive leukemia and a new promising therapeutic target.
Collapse
|
10
|
Mancini M, Leo E, Takemaru KI, Campi V, Castagnetti F, Soverini S, De Benedittis C, Rosti G, Cavo M, Santucci MA, Martinelli G. 14-3-3 Binding and Sumoylation Concur to the Down-Modulation of β-catenin Antagonist chibby 1 in Chronic Myeloid Leukemia. PLoS One 2015; 10:e0131074. [PMID: 26147002 PMCID: PMC4492953 DOI: 10.1371/journal.pone.0131074] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/28/2015] [Indexed: 11/18/2022] Open
Abstract
The down-modulation of the β-catenin antagonist Chibby 1 (CBY1) associated with the BCR-ABL1 fusion gene of chronic myeloid leukemia (CML) contributes to the aberrant activation of β-catenin, particularly in leukemic stem cells (LSC) resistant to tyrosine kinase (TK) inhibitors. It is, at least partly, driven by transcriptional events and gene promoter hyper-methylation. Here we demonstrate that it also arises from reduced protein stability upon binding to 14-3-3σ adapter protein. CBY1/14-3-3σ interaction in BCR-ABL1+ cells is mediated by the fusion protein TK and AKT phosphorylation of CBY1 at critical serine 20, and encompasses the 14-3-3σ binding modes I and II involved in the binding with client proteins. Moreover, it is impaired by c-Jun N-terminal kinase (JNK) phosphorylation of 14-3-3σ at serine 186, which promotes dissociation of client proteins. The ubiquitin proteasome system UPS participates in reducing stability of CBY1 bound with 14-3-3σ through enhanced SUMOylation. Our results open new routes towards the research on molecular pathways promoting the proliferative advantage of leukemic hematopoiesis over the normal counterpart.
Collapse
MESH Headings
- 14-3-3 Proteins/metabolism
- Amino Acid Motifs
- Benzamides/pharmacology
- Biomarkers, Tumor/metabolism
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Down-Regulation
- Exoribonucleases/metabolism
- Fusion Proteins, bcr-abl/metabolism
- Gene Expression Regulation, Leukemic/genetics
- Humans
- JNK Mitogen-Activated Protein Kinases/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Oncogene Protein p65(gag-jun)
- Phosphorylation
- Proteasome Endopeptidase Complex/metabolism
- Protein Binding
- Protein Interaction Mapping
- Protein Processing, Post-Translational
- Protein Stability
- Proto-Oncogene Proteins c-akt/metabolism
- Pyrazoles/pharmacology
- Subcellular Fractions/metabolism
- Sumoylation
- beta Catenin/antagonists & inhibitors
Collapse
Affiliation(s)
- Manuela Mancini
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
- * E-mail:
| | - Elisa Leo
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Ken-Ichi Takemaru
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York, United States of America
| | - Virginia Campi
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Fausto Castagnetti
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Simona Soverini
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Caterina De Benedittis
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Gianantonio Rosti
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Michele Cavo
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Maria Alessandra Santucci
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| | - Giovanni Martinelli
- Department of Experimental Diagnostic and Specialty Medicine—DIMES—Institute of Hematology "L. and A. Seràgnoli". University of Bologna-Medical School, Bologna, Italy
| |
Collapse
|
11
|
Tortorella SM, Hung A, Karagiannis TC. The implication of cancer progenitor cells and the role of epigenetics in the development of novel therapeutic strategies for chronic myeloid leukemia. Antioxid Redox Signal 2015; 22:1425-62. [PMID: 25366930 DOI: 10.1089/ars.2014.6096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Chronic myeloid leukemia (CML) involves the malignant transformation of hematopoietic stem cells, defined largely by the Philadelphia chromosome and expression of the breakpoint cluster region-Abelson (BCR-ABL) oncoprotein. Pharmacological tyrosine kinase inhibitors (TKIs), including imatinib mesylate, have overcome limitations in conventional treatment for the improved clinical management of CML. RECENT ADVANCES Accumulated evidence has led to the identification of a subpopulation of quiescent leukemia progenitor cells with stem-like self renewal properties that may initiate leukemogenesis, which are also shown to be present in residual disease due to their insensitivity to tyrosine kinase inhibition. CRITICAL ISSUES The characterization of quiescent leukemia progenitor cells as a unique cell population in CML pathogenesis has become critical with the complete elucidation of mechanisms involved in their survival independent of BCR-ABL that is important in the development of novel anticancer strategies. Understanding of these functional pathways in CML progenitor cells will allow for their selective therapeutic targeting. In addition, disease pathogenesis and drug responsiveness is also thought to be modulated by epigenetic regulatory mechanisms such as DNA methylation, histone acetylation, and microRNA expression, with a capacity to control CML-associated gene transcription. FUTURE DIRECTIONS A number of compounds in combination with TKIs are under preclinical and clinical investigation to assess their synergistic potential in targeting leukemic progenitor cells and/or the epigenome in CML. Despite the collective promise, further research is required in order to refine understanding, and, ultimately, advance antileukemic therapeutic strategies.
Collapse
Affiliation(s)
- Stephanie M Tortorella
- 1 Epigenomic Medicine, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct , Melbourne, Australia
| | | | | |
Collapse
|
12
|
Blanchard Z, Mullins N, Ellipeddi P, Lage JM, McKinney S, El-Etriby R, Zhang X, Isokpehi R, Hernandez B, ElShamy WM. Geminin overexpression promotes imatinib sensitive breast cancer: a novel treatment approach for aggressive breast cancers, including a subset of triple negative. PLoS One 2014; 9:e95663. [PMID: 24789045 PMCID: PMC4005756 DOI: 10.1371/journal.pone.0095663] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 03/28/2014] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is the second leading cause of cancer-related deaths in women. Triple negative breast cancer (TNBC) is an aggressive subtype that affects 10–25% mostly African American women. TNBC has the poorest prognosis of all subtypes with rapid progression leading to mortality in younger patients. So far, there is no targeted treatment for TNBC. To that end, here we show that c-Abl is one of several tyrosine kinases that phosphorylate and activate geminin’s ability to promote TNBC. Analysis of >800 breast tumor samples showed that geminin is overexpressed in ∼50% of all tumors. Although c-Abl is overexpressed in ∼90% of all tumors, it is only nuclear in geminin overexpressing tumors. In geminin-negative tumors, c-Abl is only cytoplasmic. Inhibiting c-Abl expression or activity (using imatinib or nilotinib) prevented geminin Y150 phosphorylation, inactivated the protein, and most importantly converted overexpressed geminin from an oncogene to an apoptosis inducer. In pre-clinical orthotopic breast tumor models, geminin-overexpressing cells developed aneuploid and invasive tumors, which were suppressed when c-Abl expression was blocked. Moreover, established geminin overexpressing orthotopic tumors regressed when treated with imatinib or nilotinib. Our studies support imatinib/nilotonib as a novel treatment option for patients with aggressive breast cancer (including a subset of TNBCs)-overexpressing geminin and nuclear c-Abl.
Collapse
Affiliation(s)
- Zannel Blanchard
- Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Nicole Mullins
- Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Pavani Ellipeddi
- Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Janice M. Lage
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Shawn McKinney
- Department of Surgery, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Rana El-Etriby
- Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Xu Zhang
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Raphael Isokpehi
- Center for Bioinformatics & Computational Biology, Department of Biology, Jackson State University, Jackson, Mississippi, United States of America
| | - Brenda Hernandez
- Cancer Research Center of Hawaii, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Wael M. ElShamy
- Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| |
Collapse
|
13
|
Brown S, Hutchinson CV, Aspinall-O'Dea M, Whetton AD, Johnson SM, Rees-Unwin K, Burthem J. Monocyte-derived dendritic cells from chronic myeloid leukaemia have abnormal maturation and cytoskeletal function that is associated with defective localisation and signalling by normal ABL1 protein. Eur J Haematol 2014; 93:96-102. [DOI: 10.1111/ejh.12306] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Sarah Brown
- Pharmacy Department; University of Wolverhampton; Wolverhampton UK
| | - Claire V. Hutchinson
- Institute of Cancer Sciences; Haematological Oncology; University of Manchester; Level 5 Research, St. Mary's Hospital; Manchester UK
| | - Mark Aspinall-O'Dea
- Institute of Cancer Sciences; Wolfson Molecular Imaging Centre; The University of Manchester; Manchester UK
| | - Anthony D. Whetton
- Institute of Cancer Sciences; Wolfson Molecular Imaging Centre; The University of Manchester; Manchester UK
| | - Suzanne M. Johnson
- Institute of Cancer Sciences; Manchester Academic Health Science Centre; The Christie NHS Foundation Trust; Manchester UK
| | - Karen Rees-Unwin
- Institute of Cancer Sciences; Haematological Oncology; University of Manchester; Level 5 Research, St. Mary's Hospital; Manchester UK
| | - John Burthem
- Pharmacy Department; University of Wolverhampton; Wolverhampton UK
- Clinical Haematology; Central Manchester University Hospitals; Manchester UK
| |
Collapse
|
14
|
Wnt5a enhances the response of CML cells to Imatinib Mesylate through JNK activation and γ-catenin inhibition. Leuk Res 2013; 37:1532-7. [DOI: 10.1016/j.leukres.2013.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/12/2013] [Indexed: 11/20/2022]
|
15
|
Ahmed W, Van Etten RA. Signal transduction in the chronic leukemias: implications for targeted therapies. Curr Hematol Malig Rep 2013; 8:71-80. [PMID: 23307472 DOI: 10.1007/s11899-012-0150-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The chronic leukemias, including chronic myeloid leukemia (CML), the Philadelphia-negative myeloproliferative neoplasms (MPNs), and chronic lymphocytic leukemia (CLL), have been characterized extensively for abnormalities of cellular signaling pathways. This effort has led to the elucidation of the central role of dysregulated tyrosine kinase signaling in the chronic myeloid neoplasms and of constitutive B-cell receptor signaling in CLL. This, in turn, has stimulated the development of small molecule inhibitors of these signaling pathways for therapy of chronic leukemia. Although the field is still in its infancy, the clinical results with these agents have ranged from encouraging (CLL) to spectacular (CML). In this review, we summarize recent studies that have helped to define the signaling pathways critical to the pathogenesis of the chronic leukemias. We also discuss correlative studies emerging from clinical trials of drugs targeting these pathways.
Collapse
Affiliation(s)
- Wesam Ahmed
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts Medical Center, 800 Washington Street, Box 5609, Boston, MA 02111, USA
| | | |
Collapse
|
16
|
Dual phosphorylation of Btk by Akt/protein kinase b provides docking for 14-3-3ζ, regulates shuttling, and attenuates both tonic and induced signaling in B cells. Mol Cell Biol 2013; 33:3214-26. [PMID: 23754751 DOI: 10.1128/mcb.00247-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bruton's tyrosine kinase (Btk) is crucial for B-lymphocyte activation and development. Mutations in the Btk gene cause X-linked agammaglobulinemia (XLA) in humans and X-linked immunodeficiency (Xid) in mice. Using tandem mass spectrometry, 14-3-3ζ was identified as a new binding partner and negative regulator of Btk in both B-cell lines and primary B lymphocytes. The activated serine/threonine kinase Akt/protein kinase B (PKB) phosphorylated Btk on two sites prior to 14-3-3ζ binding. The interaction sites were mapped to phosphoserine pS51 in the pleckstrin homology domain and phosphothreonine pT495 in the kinase domain. The double-alanine, S51A/T495A, replacement mutant failed to bind 14-3-3ζ, while phosphomimetic aspartate substitutions, S51D/T495D, caused enhanced interaction. The phosphatidylinositol 3-kinase (PI3-kinase) inhibitor LY294002 abrogated S51/T495 phosphorylation and binding. A newly characterized 14-3-3 inhibitor, BV02, reduced binding, as did the Btk inhibitor PCI-32765 (ibrutinib). Interestingly, in the presence of BV02, phosphorylation of Btk, phospholipase Cγ2, and NF-κB increased strongly, suggesting that 14-3-3 also regulates B-cell receptor (BCR)-mediated tonic signaling. Furthermore, downregulation of 14-3-3ζ elevated nuclear translocation of Btk. The loss-of-function mutant S51A/T495A showed reduced tyrosine phosphorylation and ubiquitination. Conversely, the gain-of-function mutant S51D/T495D exhibited intense tyrosine phosphorylation, associated with Btk ubiquitination and degradation, likely contributing to the termination of BCR signaling. Collectively, this suggests that Btk could become an important new candidate for the general study of 14-3-3-mediated regulation.
Collapse
|
17
|
Constance JE, Woessner DW, Matissek KJ, Mossalam M, Lim CS. Enhanced and selective killing of chronic myelogenous leukemia cells with an engineered BCR-ABL binding protein and imatinib. Mol Pharm 2012; 9:3318-29. [PMID: 22957899 DOI: 10.1021/mp3003539] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The oncoprotein Bcr-Abl stimulates prosurvival pathways and suppresses apoptosis from its exclusively cytoplasmic locale, but when targeted to the mitochondrial compartment of leukemia cells, Bcr-Abl was potently cytotoxic. Therefore, we designed a protein construct to act as a mitochondrial chaperone to move Bcr-Abl to the mitochondria. The chaperone (i.e., the 43.6 kDa intracellular cryptic escort (iCE)) contains an EGFP tag and two previously characterized motifs: (1) an optimized Bcr-Abl binding motif that interacts with the coiled-coil domain of Bcr (ccmut3; 72 residues), and (2) a cryptic mitochondrial targeting signal (cMTS; 51 residues) that selectively targets the mitochondria in oxidatively stressed cells (i.e., Bcr-Abl positive leukemic cells) via phosphorylation at a key residue (T193) by protein kinase C. While the iCE colocalized with Bcr-Abl, it did not relocalize to the mitochondria. However, the iCE was selectively toxic to Bcr-Abl positive K562 cells as compared to Bcr-Abl negative Cos-7 fibroblasts and 1471.1 murine breast cancer cells. The toxicity of the iCE to leukemic cells was equivalent to 10 μM imatinib at 48 h and the iCE combined with imatinib potentiated cell death beyond imatinib or the iCE alone. Substitution of either the ccmut3 or the cMTS with another Bcr-Abl binding domain (derived from Ras/Rab interaction protein 1 (RIN1; 295 residues)) or MTS (i.e., the canonical IMS derived from Smac/Diablo; 49 residues) did not match the cytotoxicity of the iCE. Additionally, a phosphorylation null mutant of the iCE also abolished the killing effect. The mitochondrial toxicity of Bcr-Abl and the iCE in Bcr-Abl positive K562 leukemia cells was confirmed by flow cytometric analysis of 7-AAD, TUNEL, and annexin-V staining. DNA segmentation and cell viability were assessed by microscopy. Subcellular localization of constructs was determined using confocal microscopy (including statistical colocalization analysis). Overall, the iCE was highly active against K562 leukemia cells and the killing effect was dependent upon both the ccmut3 and functional cMTS domains.
Collapse
Affiliation(s)
- Jonathan E Constance
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, Salt Lake City, Utah 84108, United States
| | | | | | | | | |
Collapse
|
18
|
Faley S, Copland M, Reboud J, Cooper JM. Intracellular protein trafficking kinetics in chronic myeloid leukemia stem cells using a microfluidic platform. Integr Biol (Camb) 2012; 4:368-73. [PMID: 22344285 DOI: 10.1039/c2ib00086e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The oncogenic fusion protein BCR-ABL is produced by chronic myeloid leukemia (CML) cells and functions as an abnormal, constitutively active tyrosine kinase that interferes with normal migratory and apoptotic behaviour of cells. Small molecule tyrosine kinase inhibitors (TKIs), such as dasatinib, eliminate CML progenitor cells, but fail to target the stem cell fraction resulting in persistent disease. In order to achieve a cure for CML in the majority of patients, we need an improved understanding of intracellular signalling dynamics, including the shuttling of BCR-ABL between cytosolic and nuclear compartments. In the past, the instability of BCR-ABL in assays using conventional immunohistochemical techniques has made this difficult and has not allowed for reliable analysis at the single cell level. Here we show how the utilization of rapid on-chip cell fixation within a microfluidic platform provides a means to immunofluorescently analyze the spatiotemporal localization of both BCR-ABL and c-ABL, as well as the linked apoptosis mediator, BCL-XL, in arrays of single CD34+ CML stem/progenitor cells, without cell loss. We demonstrate this proceeds up to 4 times faster than benchtop methods. Our results indicate that whilst both BCR-ABL and c-ABL shuttle from the cytoplasm to the nucleus following dasatinib treatment, the temporal dynamics are not synchronized. The microfluidic platform has the potential to provide insights into the intracellular signalling events in single cells. The ability to examine signalling events and assess BCR-ABL expression/activity in isolated cells in "real-time" may help elucidate the characteristics of rare CML stem cell events, which lead to the resistance of CML stem cells to TKIs.
Collapse
Affiliation(s)
- Shannon Faley
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37235, USA.
| | | | | | | |
Collapse
|
19
|
Computational techniques are valuable tools for the discovery of protein–protein interaction inhibitors: The 14-3-3σ case. Bioorg Med Chem Lett 2011; 21:6867-71. [DOI: 10.1016/j.bmcl.2011.09.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 09/02/2011] [Accepted: 09/03/2011] [Indexed: 12/30/2022]
|
20
|
Redig AJ, Vakana E, Platanias LC. Regulation of mammalian target of rapamycin and mitogen activated protein kinase pathways by BCR-ABL. Leuk Lymphoma 2011; 52 Suppl 1:45-53. [PMID: 21299459 DOI: 10.3109/10428194.2010.546919] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A large body of evidence has established that BCR-ABL regulates engagement and activation of mammalian target of rapamycin (mTOR) and mitogen activated protein kinase (MAPK) signaling cascades. mTOR-mediated signals, as well as signals transduced by ERK, JNK, and p38 MAPK, are important components of the aberrant signaling induced by BCR-ABL. Such deregulation of mTOR or MAPK pathways contributes to BCR-ABL leukemogenesis, and their targeting with selective inhibitors provides an approach to enhance antileukemic responses and/or overcome leukemic cell resistance in chronic myeloid leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). This review explores recent advances in our understanding of mTOR and MAPK signaling in BCR-ABL-expressing leukemias and discusses the potential therapeutic targeting of these pathways in CML and Ph+ ALL.
Collapse
Affiliation(s)
- Amanda J Redig
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Northwestern University Medical School and Jesse Brown VA Medical Center, Chicago, IL, USA
| | | | | |
Collapse
|
21
|
Mancini M, Corradi V, Petta S, Barbieri E, Manetti F, Botta M, Santucci MA. A new nonpeptidic inhibitor of 14-3-3 induces apoptotic cell death in chronic myeloid leukemia sensitive or resistant to imatinib. J Pharmacol Exp Ther 2011; 336:596-604. [PMID: 21041536 DOI: 10.1124/jpet.110.172536] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Resistance of chronic myeloid leukemia (CML) to tyrosine kinase inhibitor imatinib mesylate (IM) is most often due to point mutations in the Bcr-Abl fusion gene. T315I mutation (resulting in substitution of Ile for a Thr residue at the "gatekeeper" position 315) raises particular concern, because it also provides resistance to second-generation kinase inhibitors already approved for clinical use (nilotinib and dasatinib). Much effort is therefore focused on alternative molecular-based strategies. Previous studies proved that binding to 14-3-3 scaffolding proteins leads to cytoplasmic compartmentalization and suppression of proapoptotic and antiproliferative signals associated with Bcr-Abl protein kinase, hence contributing to leukemic clone expansion. Here we investigated the effect of 14-3-3 inhibition disruption on hematopoietic cells expressing the IM-sensitive wild type Bcr-Abl and the IM-resistant T315I mutation. Using a virtual screening protocol and docking simulations, we identified a nonpeptidic inhibitor of 14-3-3, named BV02, that exhibits a remarkable cytotoxicity against both cell types. c-Abl release from 14-3-3σ, promoting its relocation to nuclear compartment (where it triggers transcription of p73-dependent proapoptotic genes) and to mitochondrial membranes (where it induces the loss of mitochondrial transmembrane potential) combined with c-Abl enhanced association with caspase 9 (a critical step of sequential caspase activation further contributing to c-Abl pro-apoptotic function) has a prominent role in the effect of BV02 on Bcr-Abl-expressing cells. In conclusion, BV02 may be considered as a treatment option for CML and, in particular, for more advanced phases of the disease that developed IM resistance as a consequence of Bcr-Abl point mutations.
Collapse
MESH Headings
- 14-3-3 Proteins/antagonists & inhibitors
- 14-3-3 Proteins/chemistry
- Amino Acid Sequence
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/physiology
- Benzamides/chemistry
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Binding Sites/drug effects
- Binding Sites/physiology
- Cell Death/drug effects
- Cell Death/physiology
- Cell Line
- Cell Line, Tumor
- Crystallography, X-Ray
- Drug Resistance, Neoplasm/physiology
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Molecular Sequence Data
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Pyrazoles/chemistry
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
Collapse
Affiliation(s)
- Manuela Mancini
- Dipartimento di Ematologia e Scienze Oncologiche Lorenzo e Ariosto Seràgnoli, Policlinico S. Orsola, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Yamaguchi T, Miki Y, Yoshida K. The c-Abl tyrosine kinase stabilizes Pitx1 in the apoptotic response to DNA damage. Apoptosis 2010; 15:927-35. [PMID: 20563669 DOI: 10.1007/s10495-010-0488-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the DNA damage response, c-Abl tyrosine kinase is transiently accumulated in the nucleus and induces apoptosis; however, little is known about the mechanism underlying apoptosis induction via nuclear c-Abl. Here we demonstrate that the expression of human pituitary homeobox 1 (Pitx1) transcription factor is increased after DNA damage. Notably, c-Abl controls augmentation of Pitx1 at the post-transcriptional level. Overexpression of c-Abl induces tyrosine phosphorylation of Pitx1, either directly or indirectly. We also show that, upon exposure to genotoxic stress, overexpression of Pitx1 is associated with marked induction of apoptosis that is independent of p53 status. Importantly, inhibition of c-Abl kinase activity substantially attenuates Pitx1-mediated apoptosis. These findings provide evidence that c-Abl participates in modulating Pitx1 expression in the apoptotic response to DNA damage.
Collapse
Affiliation(s)
- Tomoko Yamaguchi
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | | | | |
Collapse
|
23
|
Tintori C, Veljkovic N, Veljkovic V, Botta M. Computational studies of the interaction between the HIV-1 integrase tetramer and the cofactor LEDGF/p75: Insights from molecular dynamics simulations and the Informational spectrum method. Proteins 2010; 78:3396-408. [DOI: 10.1002/prot.22847] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 07/22/2010] [Accepted: 08/03/2010] [Indexed: 01/05/2023]
|
24
|
Corradi V, Mancini M, Manetti F, Petta S, Santucci MA, Botta M. Identification of the first non-peptidic small molecule inhibitor of the c-Abl/14-3-3 protein-protein interactions able to drive sensitive and Imatinib-resistant leukemia cells to apoptosis. Bioorg Med Chem Lett 2010; 20:6133-7. [PMID: 20832303 DOI: 10.1016/j.bmcl.2010.08.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 08/03/2010] [Accepted: 08/04/2010] [Indexed: 01/11/2023]
Abstract
An in silico structure-based ligand design approach resulted in the identification of the first non-peptidic small molecule able to inhibit protein-protein interactions between 14-3-3 and c-Abl. This compound shows an anti-proliferative effect on human leukemia cells either sensitive or resistant to Imatinib, in consequence of the T315I mutation. It also mediates c-Abl release from 14-3-3 in a way similar to that found in response to Imatinib treatment.
Collapse
Affiliation(s)
- Valentina Corradi
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, Siena, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Flores O, Wang Z, Knudsen KE, Burnstein KL. Nuclear targeting of cyclin-dependent kinase 2 reveals essential roles of cyclin-dependent kinase 2 localization and cyclin E in vitamin D-mediated growth inhibition. Endocrinology 2010; 151:896-908. [PMID: 20147522 PMCID: PMC2840684 DOI: 10.1210/en.2009-1116] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
1,25-Dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)), inhibits proliferation of a variety of cell types including adenocarcinoma of the prostate. We have previously shown that 1,25-(OH)(2)D(3) increases the stability of the cyclin-dependent kinase inhibitor p27(KIP1), decreases cyclin-dependent kinase 2 (CDK2) activity, and promotes G(1) phase accumulation in human prostate cancer cells. These effects correlate with cytoplasmic relocalization of CDK2. In this study, we investigated the role of CDK2 cytoplasmic relocalization in the antiproliferative effects of 1,25-(OH)(2)D(3). CDK2 was found to be necessary for prostate cancer cell proliferation. Although induced by 1,25-(OH)(2)D(3), the cyclin-dependent kinase inhibitor p27(KIP1) was dispensable for 1,25-(OH)(2)D(3)-mediated growth inhibition. Reduction in CDK2 activity by 1,25-(OH)(2)D(3) was associated with decreased T160 phosphorylation, a residue whose phosphorylation in the nucleus is essential for CDK2 activity. Ectopic expression of cyclin E was sufficient to overcome 1,25-(OH)(2)D(3)-mediated cytoplasmic mislocalization of CDK2 and all antiproliferative effects of 1,25-(OH)(2)D(3), yet endogenous levels of cyclin E or binding to CDK2 were not affected by 1,25-(OH)(2)D(3). Similarly, knockdown of the CDK2 substrate retinoblastoma, which causes cyclin E up-regulation, resulted in resistance to 1,25-(OH)(2)D(3)-mediated growth inhibition. Human prostate cancer cells resistant to growth inhibition by 1,25-(OH)(2)D(3) but retaining fully functional vitamin D receptors were developed. These cells did not exhibit 1,25-(OH)(2)D(3)-mediated cytoplasmic relocalization of CDK2. Targeting CDK2 to the nucleus of 1,25-(OH)(2)D(3)-sensitive cancer cells blocked G(1) accumulation and growth inhibition by 1,25-(OH)(2)D(3). These data establish central roles for CDK2 nuclear-cytoplasmic trafficking and cyclin E in the mechanism of 1,25-(OH)(2)D(3)-mediated growth inhibition in prostate cancer cells.
Collapse
Affiliation(s)
- Omar Flores
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Miami, Florida 33136, USA
| | | | | | | |
Collapse
|
26
|
Mancini M, Corradi V, Petta S, Martinelli G, Barbieri E, Santucci MA. mTOR inhibitor RAD001 (Everolimus) enhances the effects of imatinib in chronic myeloid leukemia by raising the nuclear expression of c-ABL protein. Leuk Res 2009; 34:641-8. [PMID: 19643477 DOI: 10.1016/j.leukres.2009.07.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 07/01/2009] [Accepted: 07/04/2009] [Indexed: 01/21/2023]
Abstract
Constitutive tyrosine kinase (TK) activity of p210 BCR-ABL fusion protein of chronic myeloid leukemia (CML) usurps physiological functions of normal p145 c-ABL protein. Accordingly, its inhibition by imatinib mesylate (IM) lets p145 c-ABL translocate into the nuclear compartment, which drives cell growth arrest and apoptotic death. Here we show that IM and the mammalian target of rapamycin (mTOR) inhibitor RAD001 (Everolimus) have additive effects on BCR-ABL-expressing cells. Those effects are at least partly conditional upon the enhanced nuclear accumulation of p145 c-ABL through events encompassing post-translational modifications of p145 c-ABL (Thr(735) phosphorylation) precluding its nuclear export and of 14-3-3 sigma (Ser(186) phosphorylation by c-Jun N-terminal kinase [JNK]) promoting p145 c-ABL nuclear re-import.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Benzamides
- Blotting, Western
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cell Separation
- Everolimus
- Flow Cytometry
- Gene Expression/drug effects
- Humans
- Imatinib Mesylate
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Microscopy, Confocal
- Piperazines/administration & dosage
- Protein Processing, Post-Translational/drug effects
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Transport/drug effects
- Proto-Oncogene Proteins c-abl/biosynthesis
- Proto-Oncogene Proteins c-abl/drug effects
- Pyrimidines/administration & dosage
- Sirolimus/administration & dosage
- Sirolimus/analogs & derivatives
- TOR Serine-Threonine Kinases
Collapse
Affiliation(s)
- Manuela Mancini
- Dipartimento di Ematologia e Scienze Oncologiche Lorenzo e Ariosto Seràgnoli, University of Bologna, Italy.
| | | | | | | | | | | |
Collapse
|