1
|
Rout AK, Dehury B, Parida SN, Rout SS, Jena R, Kaushik N, Kaushik NK, Pradhan SK, Sahoo CR, Singh AK, Arya M, Behera BK. A review on structure-function mechanism and signaling pathway of serine/threonine protein PIM kinases as a therapeutic target. Int J Biol Macromol 2024; 270:132030. [PMID: 38704069 DOI: 10.1016/j.ijbiomac.2024.132030] [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: 11/24/2023] [Revised: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
The proviral integration for the Moloney murine leukemia virus (PIM) kinases, belonging to serine/threonine kinase family, have been found to be overexpressed in various types of cancers, such as prostate, breast, colon, endometrial, gastric, and pancreatic cancer. The three isoforms PIM kinases i.e., PIM1, PIM2, and PIM3 share a high degree of sequence and structural similarity and phosphorylate substrates controlling tumorigenic phenotypes like proliferation and cell survival. Targeting short-lived PIM kinases presents an intriguing strategy as in vivo knock-down studies result in non-lethal phenotypes, indicating that clinical inhibition of PIM might have fewer adverse effects. The ATP binding site (hinge region) possesses distinctive attributes, which led to the development of novel small molecule scaffolds that target either one or all three PIM isoforms. Machine learning and structure-based approaches have been at the forefront of developing novel and effective chemical therapeutics against PIM in preclinical and clinical settings, and none have yet received approval for cancer treatment. The stability of PIM isoforms is maintained by PIM kinase activity, which leads to resistance against PIM inhibitors and chemotherapy; thus, to overcome such effects, PIM proteolysis targeting chimeras (PROTACs) are now being developed that specifically degrade PIM proteins. In this review, we recapitulate an overview of the oncogenic functions of PIM kinases, their structure, function, and crucial signaling network in different types of cancer, and the potential of pharmacological small-molecule inhibitors. Further, our comprehensive review also provides valuable insights for developing novel antitumor drugs that specifically target PIM kinases in the future. In conclusion, we provide insights into the benefits of degrading PIM kinases as opposed to blocking their catalytic activity to address the oncogenic potential of PIM kinases.
Collapse
Affiliation(s)
- Ajaya Kumar Rout
- Rani Lakshmi Bai Central Agricultural University, Jhansi-284003, Uttar Pradesh, India
| | - Budheswar Dehury
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal-576104, India
| | - Satya Narayan Parida
- Rani Lakshmi Bai Central Agricultural University, Jhansi-284003, Uttar Pradesh, India
| | - Sushree Swati Rout
- Department of Zoology, Fakir Mohan University, Balasore-756089, Odisha, India
| | - Rajkumar Jena
- Department of Zoology, Fakir Mohan University, Balasore-756089, Odisha, India
| | - Neha Kaushik
- Department of Biotechnology, The University of Suwon, Hwaseong si, South Korea
| | | | - Sukanta Kumar Pradhan
- Department of Bioinformatics, Odisha University of Agriculture and Technology, Bhubaneswar-751003, Odisha, India
| | - Chita Ranjan Sahoo
- ICMR-Regional Medical Research Centre, Department of Health Research, Ministry of Health and Family Welfare, Government of India, Bhubaneswar-751023, India
| | - Ashok Kumar Singh
- Rani Lakshmi Bai Central Agricultural University, Jhansi-284003, Uttar Pradesh, India
| | - Meenakshi Arya
- Rani Lakshmi Bai Central Agricultural University, Jhansi-284003, Uttar Pradesh, India.
| | - Bijay Kumar Behera
- Rani Lakshmi Bai Central Agricultural University, Jhansi-284003, Uttar Pradesh, India.
| |
Collapse
|
2
|
Lagunas-Rangel FA. DNA damage accumulation and repair defects in FLT3-ITD acute myeloid leukemia: Implications for clonal evolution and disease progression. Hematol Oncol 2023; 41:26-38. [PMID: 36131612 PMCID: PMC10087755 DOI: 10.1002/hon.3076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/15/2022] [Accepted: 09/17/2022] [Indexed: 02/03/2023]
Abstract
Acute myeloid leukemia is a group of hematological diseases that have a high mortality rate. During the development of this pathology, hematopoietic cells acquire chromosomal rearrangements and multiple genetic mutations, including FLT3-ITD. FLT3-ITD is a marker associated with a poor clinical prognosis and involves the activation of pathways such as PI3K/AKT, MAPK/ERK, and JAK/STAT that favor the survival and proliferation of leukemic cells. In addition, FLT3-ITD leads to overproduction of reactive oxygen species and defective DNA damage repair, both implicated in the appearance of new mutations and leukemic clones. Thus, the purpose of this review is to illustrate the molecular mechanisms through which FLT3-ITD generates genetic instability and how it facilitates clonal evolution with the generation of more resistant and aggressive cells. Likewise, this article discusses the feasibility of combined therapies with FLT3 inhibitors and inhibitors of DNA repair pathways.
Collapse
|
3
|
Zhang Y, Yuan L. Fms-like tyrosine kinase 3-internal tandem duplications epigenetically activates checkpoint kinase 1 in acute myeloid leukemia cells. Sci Rep 2021; 11:13236. [PMID: 34168220 PMCID: PMC8225911 DOI: 10.1038/s41598-021-92566-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/04/2021] [Indexed: 01/15/2023] Open
Abstract
It is not clear how Fms-like tyrosine kinase 3-internal tandem duplications (FLT3-ITD) regulates checkpoint kinase 1 (CHK1) in acute myeloid leukemia (AML). In this study, we investigated the regulatory effect of FLT3-ITD on CHK1. Our results showed that CHK1 was highly expressed in FLT3-ITD positive AML. The overall survival rate and disease-free survival rate of AML patients with high CHK1 level were lower than those of patients with low CHK1 level. Mechanistically, FLT3-ITD recruited p300 to the CHK1 promoter and subsequently acetylated H3K27, thereby enhancing the transcription of CHK1. Interfering with the expression of CHK1 significantly inhibited the cell proliferation and induced cell apoptosis in FLT3-ITD positive MV4-11 cells. In addition, CHK1 knockdown promoted the sensitivity of MV4-11 cells to the epigenetic inhibitors JQ1 and C646. This study discovers a new therapeutic target for FLT3-ITD + AML and provided evidence for the combination of epigenetic inhibitors for AML treatment.
Collapse
Affiliation(s)
- Yudong Zhang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Lingli Yuan
- Department of Hematology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
4
|
Wang Z, Cai J, Cheng J, Yang W, Zhu Y, Li H, Lu T, Chen Y, Lu S. FLT3 Inhibitors in Acute Myeloid Leukemia: Challenges and Recent Developments in Overcoming Resistance. J Med Chem 2021; 64:2878-2900. [PMID: 33719439 DOI: 10.1021/acs.jmedchem.0c01851] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene are often present in newly diagnosed acute myeloid leukemia (AML) patients with an incidence rate of approximately 30%. Recently, many FLT3 inhibitors have been developed and exhibit positive preclinical and clinical effects against AML. However, patients develop resistance soon after undergoing FLT3 inhibitor treatment, resulting in short durable responses and poor clinical effects. This review will discuss the main mechanisms of resistance to clinical FLT3 inhibitors and summarize the emerging strategies that are utilized to overcome drug resistance. Basically, medicinal chemistry efforts to develop new small-molecule FLT3 inhibitors offer a direct solution to this problem. Other potential strategies include the combination of FLT3 inhibitors with other therapies and the development of multitarget inhibitors. It is hoped that this review will provide inspiring insights into the discovery of new AML therapies that can eventually overcome the resistance to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Wenqianzi Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
5
|
Neizer-Ashun F, Bhattacharya R. Reality CHEK: Understanding the biology and clinical potential of CHK1. Cancer Lett 2020; 497:202-211. [PMID: 32991949 DOI: 10.1016/j.canlet.2020.09.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/26/2020] [Accepted: 09/20/2020] [Indexed: 12/13/2022]
Abstract
The DNA damage response enables cells to cope with various stresses that threaten genomic integrity. A critical component of this response is the serine/threonine kinase CHK1 which is encoded by the CHEK1 gene. Originally identified as a regulator of the G2/M checkpoint, CHK1 has since been shown to play important roles in DNA replication, mitotic progression, DNA repair, and overall cell cycle regulation. However, the potential of CHK1 as a cancer therapy has not been realized clinically. Herein we expound our current understanding of the principal roles of CHK1 and highlight different avenues for CHK1 targeting in cancer therapy.
Collapse
Affiliation(s)
- Fiifi Neizer-Ashun
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, United States
| | - Resham Bhattacharya
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, United States; Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, United States; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
6
|
Santio NM, Vainio V, Hoikkala T, Mung KL, Lång M, Vahakoski R, Zdrojewska J, Coffey ET, Kremneva E, Rainio EM, Koskinen PJ. PIM1 accelerates prostate cancer cell motility by phosphorylating actin capping proteins. Cell Commun Signal 2020; 18:121. [PMID: 32771000 PMCID: PMC7414696 DOI: 10.1186/s12964-020-00618-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/27/2020] [Indexed: 12/22/2022] Open
Abstract
Background The PIM family kinases promote cancer cell survival and motility as well as metastatic growth in various types of cancer. We have previously identified several PIM substrates, which support cancer cell migration and invasiveness. However, none of them are known to regulate cellular movements by directly interacting with the actin cytoskeleton. Here we have studied the phosphorylation-dependent effects of PIM1 on actin capping proteins, which bind as heterodimers to the fast-growing actin filament ends and stabilize them. Methods Based on a phosphoproteomics screen for novel PIM substrates, we have used kinase assays and fluorescence-based imaging techniques to validate actin capping proteins as PIM1 substrates and interaction partners. We have analysed the functional consequences of capping protein phosphorylation on cell migration and adhesion by using wound healing and real-time impedance-based assays. We have also investigated phosphorylation-dependent effects on actin polymerization by analysing the protective role of capping protein phosphomutants in actin disassembly assays. Results We have identified capping proteins CAPZA1 and CAPZB2 as PIM1 substrates, and shown that phosphorylation of either of them leads to increased adhesion and migration of human prostate cancer cells. Phosphorylation also reduces the ability of the capping proteins to protect polymerized actin from disassembly. Conclusions Our data suggest that PIM kinases are able to induce changes in actin dynamics to support cell adhesion and movement. Thus, we have identified a novel mechanism through which PIM kinases enhance motility and metastatic behaviour of cancer cells. Video abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Niina M Santio
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Veera Vainio
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Tuuli Hoikkala
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Kwan Long Mung
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Mirka Lång
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Riitta Vahakoski
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Justyna Zdrojewska
- Turku Bioscience, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Eleanor T Coffey
- Turku Bioscience, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Elena Kremneva
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Eeva-Marja Rainio
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Päivi J Koskinen
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland.
| |
Collapse
|
7
|
Li Y, Cheng Y, Zhang M, He X, Kong L, Zhou K, Zhou Y, Li L, Tian H, Song X, Cui Y. A New Compound with Increased Antitumor Activity by Cotargeting MEK and Pim-1. iScience 2020; 23:101254. [PMID: 32585592 PMCID: PMC7322072 DOI: 10.1016/j.isci.2020.101254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 02/05/2023] Open
Abstract
Feedback circuits are one of the major causes underlying tumor resistance. Thus, compounds that target one oncogenic pathway with simultaneously blocking its compensatory pathway will be of great value for cancer treatment. Here, we develop a new MEK inhibitor designated as KZ-02 that exhibits unexpectedly higher cytotoxicity than its starting compound AZD6244, a well-known MEK inhibitor, in colorectal cancer (CRC). Subsequent kinase selectivity study identified Pim-1 as an additional cellular target for KZ-02. Further studies showed that AZD6244 and Pim-1 1 (a Pim-1 inhibitor) have a synergistic effect on CRC suppression. Mechanistic study revealed that MEK inhibition by AZD6244 leads to increased Pim-1 expression, which could be a general mechanism behind the compromised cell-killing activity of MEK inhibitors. KZ-02, despite increasing Pim-1 mRNA expression, simultaneously promotes Pim-1 proteasomal degradation. Therefore, we uncover a new MEK inhibitor KZ-02 with significantly enhanced antitumor activity by co-targeting MEK and Pim-1.
Collapse
Affiliation(s)
- Yanan Li
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou University Medical College Cancer Hospital, 7 Raoping Road, Shantou, Guangdong 515031, China
| | - Ying Cheng
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Maoqi Zhang
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou University Medical College Cancer Hospital, 7 Raoping Road, Shantou, Guangdong 515031, China
| | - Xiaoli He
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Li Kong
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou University Medical College Cancer Hospital, 7 Raoping Road, Shantou, Guangdong 515031, China
| | - Kexiang Zhou
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou University Medical College Cancer Hospital, 7 Raoping Road, Shantou, Guangdong 515031, China
| | - Yunfu Zhou
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Lin Li
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hongqi Tian
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China.
| | - Xiaomin Song
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Yukun Cui
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou University Medical College Cancer Hospital, 7 Raoping Road, Shantou, Guangdong 515031, China.
| |
Collapse
|
8
|
Long L, Assaraf YG, Lei ZN, Peng H, Yang L, Chen ZS, Ren S. Genetic biomarkers of drug resistance: A compass of prognosis and targeted therapy in acute myeloid leukemia. Drug Resist Updat 2020; 52:100703. [PMID: 32599434 DOI: 10.1016/j.drup.2020.100703] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy with complex heterogenous genetic and biological nature. Thus, prognostic prediction and targeted therapies might contribute to better chemotherapeutic response. However, the emergence of multidrug resistance (MDR) markedly impedes chemotherapeutic efficacy and dictates poor prognosis. Therefore, prior evaluation of chemoresistance is of great importance in therapeutic decision making and prognosis. In recent years, preclinical studies on chemoresistance have unveiled a compendium of underlying molecular basis, which facilitated the development of targetable small molecules. Furthermore, routing genomic sequencing has identified various genomic aberrations driving cellular response during the course of therapeutic treatment through adaptive mechanisms of drug resistance, some of which serve as prognostic biomarkers in risk stratification. However, the underlying mechanisms of MDR have challenged the certainty of the prognostic significance of some mutations. This review aims to provide a comprehensive understanding of the role of MDR in therapeutic decision making and prognostic prediction in AML. We present an updated genetic landscape of the predominant mechanisms of drug resistance with novel targeted therapies and potential prognostic biomarkers from preclinical and clinical chemoresistance studies in AML. We particularly highlight the unfolded protein response (UPR) that has emerged as a critical regulatory pathway in chemoresistance of AML with promising therapeutic horizon. Futhermore, we outline the most prevalent mutations associated with mechanisms of chemoresistance and delineate the future directions to improve the current prognostic tools. The molecular analysis of chemoresistance integrated with genetic profiling will facilitate decision making towards personalized prognostic prediction and enhanced therapeutic efficacy.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Disease-Free Survival
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Multiple/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Molecular Targeted Therapy/methods
- Mutation
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/prevention & control
- Precision Medicine/methods
- Prognosis
- Unfolded Protein Response/genetics
Collapse
Affiliation(s)
- Luyao Long
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China; Graduate School, Chinese Academy of Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zi-Ning Lei
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA; School of Public Health, Guangzhou Medical University, Guangzhou, P.R. China
| | - Hongwei Peng
- Department of Pharmacy, First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Lin Yang
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Simei Ren
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China; Graduate School, Chinese Academy of Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China; Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China.
| |
Collapse
|
9
|
All-trans retinoic acid exerts selective anti-FLT3-ITD acute myeloid leukemia efficacy through downregulating Chk1 kinase. Cancer Lett 2020; 473:130-138. [PMID: 31904486 DOI: 10.1016/j.canlet.2019.12.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/27/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
All-trans retinoic acid (ATRA) is known to be a potent inhibitor of FLT3-ITD acute myeloid leukemia (AML) cells, although the exact mechanism remains unclear. In this work, we report that ATRA causes fatal mitotic catastrophe in FLT3-ITD AML cells by degrading Chk1 kinase, and therefore preventing DNA damage repair. In order to explore a further enhancement in the inhibitory effect of ATRA on FLT3-ITD AML cells, we investigated the suitability of a combination of ATRA and DNA damage drug SN38. In vitro experiments showed that this combinatorial approach effectively inhibited the proliferation of FLT3-ITD cells and induced cell apoptosis in AML. In vivo experiments confirmed that the combination could substantially improve the anti-tumor effect of SN38. Taken together, our results indicate that ATRA down-regulates Chk1 in FLT3-ITD AML cells, and the combination of ATRA and SN38 significantly improves the anti-tumor effect of either ATRA or SN38 when used alone.
Collapse
|
10
|
Multifaceted Regulation of PTEN Subcellular Distributions and Biological Functions. Cancers (Basel) 2019; 11:cancers11091247. [PMID: 31454965 PMCID: PMC6770588 DOI: 10.3390/cancers11091247] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/15/2019] [Accepted: 08/19/2019] [Indexed: 12/19/2022] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor gene frequently found to be inactivated in over 30% of human cancers. PTEN encodes a 54-kDa lipid phosphatase that serves as a gatekeeper of the phosphoinositide 3-kinase pathway involved in the promotion of multiple pro-tumorigenic phenotypes. Although the PTEN protein plays a pivotal role in carcinogenesis, cumulative evidence has implicated it as a key signaling molecule in several other diseases as well, such as diabetes, Alzheimer's disease, and autism spectrum disorders. This finding suggests that diverse cell types, especially differentiated cells, express PTEN. At the cellular level, PTEN is widely distributed in all subcellular compartments and organelles. Surprisingly, the cytoplasmic compartment, not the plasma membrane, is the predominant subcellular location of PTEN. More recently, the finding of a secreted 'long' isoform of PTEN and the presence of PTEN in the cell nucleus further revealed unexpected biological functions of this multifaceted molecule. At the regulatory level, PTEN activity, stability, and subcellular distribution are modulated by a fascinating array of post-translational modification events, including phosphorylation, ubiquitination, and sumoylation. Dysregulation of these regulatory mechanisms has been observed in various human diseases. In this review, we provide an up-to-date overview of the knowledge gained in the last decade on how different functional domains of PTEN regulate its biological functions, with special emphasis on its subcellular distribution. This review also highlights the findings of published studies that have reported how mutational alterations in specific PTEN domains can lead to pathogenesis in humans.
Collapse
|
11
|
Chen J, Tang G. PIM-1 kinase: a potential biomarker of triple-negative breast cancer. Onco Targets Ther 2019; 12:6267-6273. [PMID: 31496730 PMCID: PMC6690594 DOI: 10.2147/ott.s212752] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/30/2019] [Indexed: 01/10/2023] Open
Abstract
Triple-negative breast cancer is associated with a poor prognosis, and effective biomarkers for targeted diagnosis and treatment are lacking. The tumorigenicity of the provirus integration site for Moloney murine leukemia virus 1 (PIM-1) gene has been studied for many years. However, its significance in breast cancer remains unclear. In this review we briefly summarized the physiological characteristics and regulation of PIM-1 kinase, and subsequently focused on the role of PIM-1 in tumors, especially breast cancer. Oncogene PIM-1 was found to be upregulated in breast cancer, especially in triple-negative breast cancer. Moreover, it is involved in tumorigenesis and the development of drug resistance, and linked to poor prognosis. A highly selective probe targeting PIM-1 for imaging has emerged, suggesting that PIM-1 may be a potential biomarker for the accurate diagnosis and targeted therapy of triple-negative breast cancer.
Collapse
Affiliation(s)
- Jieying Chen
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Guangyu Tang
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Cervantes-Gomez F, Stellrecht CM, Ayres ML, Keating MJ, Wierda WG, Gandhi V. PIM kinase inhibitor, AZD1208, inhibits protein translation and induces autophagy in primary chronic lymphocytic leukemia cells. Oncotarget 2019; 10:2793-2809. [PMID: 31073371 PMCID: PMC6497463 DOI: 10.18632/oncotarget.26876] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/23/2019] [Indexed: 11/25/2022] Open
Abstract
The PIM1, PIM2, and PIM3 serine/threonine kinases play a role in the proliferation and survival of cancer cells. Mice lacking these three kinases were viable. Further, in human hematological malignancies, these proteins are overexpressed making them suitable targets. Several small molecule inhibitors against this enzyme were synthesized and tested. AZD1208, an orally available small-molecule drug, inhibits all three PIM kinases at a low nanomolar range. AZD1208 has been tested in clinical trials for patients with solid tumors and hematological malignancies, especially acute myelogenous leukemia. The present study evaluated the efficacy and biological actions of AZD1208 in chronic lymphocytic leukemia (CLL) cells. CLL cells had higher levels of PIM2 protein and mRNAs than did normal lymphocytes from healthy donors. Treatment of CLL lymphocytes with AZD1208 resulted in modest cell death, whereas practically no cytotoxicity was observed in healthy lymphocytes. To determine the mechanism by which AZD1208 inhibits PIM kinase function, we evaluated PIM kinase pathway and downstream substrates. Because peripheral blood CLL cells are replicationally quiescent, we analyzed substrates involved in apoptosis, transcription, and translation but not cell cycle targets. AZD1208 inhibited protein translation by decreasing phosphorylation levels of 4E-binding protein 1 (4E-BP1). AZD1208 induced autophagy in replicationally-quiescent CLL cells, which is consistent with protein translation inhibition. These data suggest that AZD1208 may elicit cytotoxicity in CLL cells through inhibiting translation and autophagy induction.
Collapse
Affiliation(s)
- Fabiola Cervantes-Gomez
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine M Stellrecht
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX, USA
| | - Mary L Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
13
|
Wu M, Li L, Hamaker M, Small D, Duffield AS. FLT3-ITD cooperates with Rac1 to modulate the sensitivity of leukemic cells to chemotherapeutic agents via regulation of DNA repair pathways. Haematologica 2019; 104:2418-2428. [PMID: 30975911 PMCID: PMC6959181 DOI: 10.3324/haematol.2018.208843] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 04/09/2019] [Indexed: 01/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm, and patients with an internal tandem duplication (ITD) mutation of the FMS-like tyrosine kinase-3 (FLT3) receptor gene have a poor prognosis. FLT3-ITD interacts with DOCK2, a G effector protein that activates Rac1/2. Previously, we showed that knockdown of DOCK2 leads to decreased survival of FLT3-ITD leukemic cells. We further investigated the mechanisms by which Rac1/DOCK2 activity affects cell survival and chemotherapeutic response in FLT3-ITD leukemic cells. Exogenous expression of FLT3-ITD led to increased Rac1 activity, reactive oxygen species, phosphorylated STAT5, DNA damage response factors and cytarabine resistance. Conversely, DOCK2 knockdown resulted in a decrease in these factors. Consistent with the reduction in DNA damage response factors, FLT3-ITD cells with DOCK2 knockdown exhibited significantly increased sensitivity to DNA damage response inhibitors. Moreover, in a mouse model of FLT3-ITD AML, animals treated with the CHK1 inhibitor MK8776 + cytarabine survived longer than those treated with cytarabine alone. These findings suggest that FLT3-ITD and Rac1 activity cooperatively modulate DNA repair activity, the addition of DNA damage response inhibitors to conventional chemotherapy may be useful in the treatment of FLT3-ITD AML, and inhibition of the Rac signaling pathways via DOCK2 may provide a novel and promising therapeutic target for FLT3-ITD AML.
Collapse
Affiliation(s)
| | - Li Li
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | | - Donald Small
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | |
Collapse
|
14
|
Adam K, Cartel M, Lambert M, David L, Yuan L, Besson A, Mayeux P, Manenti S, Didier C. A PIM-CHK1 signaling pathway regulates PLK1 phosphorylation and function during mitosis. J Cell Sci 2018; 131:jcs213116. [PMID: 29976560 DOI: 10.1242/jcs.213116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 06/15/2018] [Indexed: 11/20/2022] Open
Abstract
Although the kinase CHK1 is a key player in the DNA damage response (DDR), several studies have recently provided evidence of DDR-independent roles of CHK1, in particular following phosphorylation of its S280 residue. Here, we demonstrate that CHK1 S280 phosphorylation is cell cycle-dependent and peaks during mitosis. We found that this phosphorylation was catalyzed by the kinase PIM2, whose protein expression was also increased during mitosis. Importantly, we identified polo-like kinase 1 (PLK1) as a direct target of CHK1 during mitosis. Genetic or pharmacological inhibition of CHK1 reduced the activating phosphorylation of PLK1 on T210, and recombinant CHK1 was able to phosphorylate T210 of PLK1 in vitro Accordingly, S280-phosphorylated CHK1 and PLK1 exhibited similar specific mitotic localizations, and PLK1 was co-immunoprecipitated with S280-phosphorylated CHK1 from mitotic cell extracts. Moreover, CHK1-mediated phosphorylation of PLK1 was dependent on S280 phosphorylation by PIM2. Inhibition of PIM proteins reduced cell proliferation and mitotic entry, which was rescued by expressing a T210D phosphomimetic mutant of PLK1. Altogether, these data identify a new PIM-CHK1-PLK1 phosphorylation cascade that regulates different mitotic steps independently of the CHK1 DDR function.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Kévin Adam
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, 75014 Paris, France
- Ligue Nationale Contre le Cancer, équipe labellisée
| | - Maëlle Cartel
- Ligue Nationale Contre le Cancer, équipe labellisée
- Cancer Research Center of Toulouse, INSERM U1037, CNRS ERL 5294, Université de Toulouse, 31100 Toulouse, France
| | - Mireille Lambert
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, 75014 Paris, France
- Ligue Nationale Contre le Cancer, équipe labellisée
| | - Laure David
- Ligue Nationale Contre le Cancer, équipe labellisée
- Cancer Research Center of Toulouse, INSERM U1037, CNRS ERL 5294, Université de Toulouse, 31100 Toulouse, France
| | - Lingli Yuan
- Department of Hematology, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Furong, Changsha, Hunan 410011, China
| | - Arnaud Besson
- Cancer Research Center of Toulouse, INSERM U1037, CNRS ERL 5294, Université de Toulouse, 31100 Toulouse, France
| | - Patrick Mayeux
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, 75014 Paris, France
- Ligue Nationale Contre le Cancer, équipe labellisée
| | - Stéphane Manenti
- Ligue Nationale Contre le Cancer, équipe labellisée
- Cancer Research Center of Toulouse, INSERM U1037, CNRS ERL 5294, Université de Toulouse, 31100 Toulouse, France
| | - Christine Didier
- Ligue Nationale Contre le Cancer, équipe labellisée
- Cancer Research Center of Toulouse, INSERM U1037, CNRS ERL 5294, Université de Toulouse, 31100 Toulouse, France
| |
Collapse
|
15
|
Santio NM, Koskinen PJ. PIM kinases: From survival factors to regulators of cell motility. Int J Biochem Cell Biol 2017; 93:74-85. [DOI: 10.1016/j.biocel.2017.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 01/01/2023]
|
16
|
Rebello RJ, Huglo AV, Furic L. PIM activity in tumours: A key node of therapy resistance. Adv Biol Regul 2017; 67:163-169. [PMID: 29111105 DOI: 10.1016/j.jbior.2017.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/20/2017] [Accepted: 10/20/2017] [Indexed: 10/18/2022]
Abstract
The PIM kinases are proto-oncogenes which have been shown to facilitate cell survival and proliferation to drive malignancy and resistance post-therapy. They are able to suppress cell death signals, sustain PI3K/AKT/mTORC1 pathway activity and regulate the MYC oncogenic program. Recent work has revealed PIM kinase essentiality for advanced tumour maintenance and described tumour sensitivity to small molecule inhibitors targeting PIM kinase in multiple malignancies.
Collapse
Affiliation(s)
- Richard J Rebello
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, VIC, 3800, Australia
| | - Alisée V Huglo
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Luc Furic
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, VIC, 3800, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
17
|
Silverbush D, Grosskurth S, Wang D, Powell F, Gottgens B, Dry J, Fisher J. Cell-Specific Computational Modeling of the PIM Pathway in Acute Myeloid Leukemia. Cancer Res 2017; 77:827-838. [PMID: 27965317 DOI: 10.1158/0008-5472.can-16-1578] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/09/2016] [Accepted: 11/30/2016] [Indexed: 11/16/2022]
Abstract
Personalized therapy is a major goal of modern oncology, as patient responses vary greatly even within a histologically defined cancer subtype. This is especially true in acute myeloid leukemia (AML), which exhibits striking heterogeneity in molecular segmentation. When calibrated to cell-specific data, executable network models can reveal subtle differences in signaling that help explain differences in drug response. Furthermore, they can suggest drug combinations to increase efficacy and combat acquired resistance. Here, we experimentally tested dynamic proteomic changes and phenotypic responses in diverse AML cell lines treated with pan-PIM kinase inhibitor and fms-related tyrosine kinase 3 (FLT3) inhibitor as single agents and in combination. We constructed cell-specific executable models of the signaling axis, connecting genetic aberrations in FLT3, tyrosine kinase 2 (TYK2), platelet-derived growth factor receptor alpha (PDGFRA), and fibroblast growth factor receptor 1 (FGFR1) to cell proliferation and apoptosis via the PIM and PI3K kinases. The models capture key differences in signaling that later enabled them to accurately predict the unique proteomic changes and phenotypic responses of each cell line. Furthermore, using cell-specific models, we tailored combination therapies to individual cell lines and successfully validated their efficacy experimentally. Specifically, we showed that cells mildly responsive to PIM inhibition exhibited increased sensitivity in combination with PIK3CA inhibition. We also used the model to infer the origin of PIM resistance engineered through prolonged drug treatment of MOLM16 cell lines and successfully validated experimentally our prediction that this resistance can be overcome with AKT1/2 inhibition. Cancer Res; 77(4); 827-38. ©2016 AACR.
Collapse
Affiliation(s)
- Dana Silverbush
- Department of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
- Microsoft Research, Cambridge, UK
| | | | | | | | - Berthold Gottgens
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust and MRC Stem Cell Institute, University of Cambridge, UK
| | - Jonathan Dry
- AstraZeneca Oncology IMED, Waltham, Massachusetts.
| | - Jasmin Fisher
- Microsoft Research, Cambridge, UK.
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Wu J, Zhang Q, Wuu YR, Zou S, Hei TK. Cytoplasmic Irradiation Induces Metabolic Shift in Human Small Airway Epithelial Cells via Activation of Pim-1 Kinase. Radiat Res 2017; 187:441-453. [PMID: 28170315 DOI: 10.1667/rr0006cc.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The unique cellular and molecular consequences of cytoplasmic damage caused by ionizing radiation were studied using a precision microbeam irradiator. Our results indicated that targeted cytoplasmic irradiation induced metabolic shift from an oxidative to glycolytic phenotype in human small airway epithelial cells (SAE). At 24 h postirradiation, there was an increase in the mRNA expression level of key glycolytic enzymes as well as lactate secretion in SAE cells. Using RNA-sequencing analysis to compare genes that were responsive to cytoplasmic versus nuclear irradiation, we found a glycolysis related gene, Pim-1, was significantly upregulated only in cytoplasmic irradiated SAE cells. Inhibition of Pim-1 activity using the selective pharmaceutic inhibitor Smi-4a significantly reduced the level of lactate production and glucose uptake after cytoplasmic irradiation. In addition, Pim-1 also inhibited AMPK activity, which is a well-characterized negative regulator of glycolysis. Distinct from the glycolysis induced by cytoplasmic irradiation, targeted nuclear irradiation also induced a transient and minimal increase in glycolysis that correlated with increased expression of Hif-1α. In an effort to explore the underline mechanism, we found that inhibition of mitochondria fission using the cell-permeable inhibitor mdivi-1 suppressed the induction of Pim-1, thus confirming Pim-1 upregulation as a downstream effect of mitochondrial dysfunction. Our data show and, for the first time, that cytoplasmic irradiation mediate expression level of Pim-1, which lead to glycolytic shift in SAE cells. Additionally, since glycolysis is frequently linked to cancer cell metabolism, our findings further suggest a role of cytoplasmic damage in promoting neoplastic changes.
Collapse
Affiliation(s)
- Jinhua Wu
- a Center for Radiological Research, College of Physicians and Surgeons; Columbia University, New York, New York 10032.,d Institute of Plasma Physics, Chinese Academy of Sciences, Hefei, Anhui, China 230031
| | - Qin Zhang
- c Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University. Chengdu, Sichuan, China 610041; and
| | - Yen-Ruh Wuu
- a Center for Radiological Research, College of Physicians and Surgeons; Columbia University, New York, New York 10032
| | - Sirui Zou
- a Center for Radiological Research, College of Physicians and Surgeons; Columbia University, New York, New York 10032
| | - Tom K Hei
- a Center for Radiological Research, College of Physicians and Surgeons; Columbia University, New York, New York 10032.,b Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| |
Collapse
|
19
|
Bertoli S, Boutzen H, David L, Larrue C, Vergez F, Fernandez-Vidal A, Yuan L, Hospital MA, Tamburini J, Demur C, Delabesse E, Saland E, Sarry JE, Galcera MO, Mansat-De Mas V, Didier C, Dozier C, Récher C, Manenti S. CDC25A governs proliferation and differentiation of FLT3-ITD acute myeloid leukemia. Oncotarget 2016; 6:38061-78. [PMID: 26515730 PMCID: PMC4741984 DOI: 10.18632/oncotarget.5706] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/06/2015] [Indexed: 11/25/2022] Open
Abstract
We investigated cell cycle regulation in acute myeloid leukemia cells expressing the FLT3-ITD mutated tyrosine kinase receptor, an underexplored field in this disease. Upon FLT3 inhibition, CDC25A mRNA and protein were rapidly down-regulated, while levels of other cell cycle proteins remained unchanged. This regulation was dependent on STAT5, arguing for FLT3-ITD-dependent transcriptional regulation of CDC25A. CDC25 inhibitors triggered proliferation arrest and cell death of FLT3-ITD as well as FLT3-ITD/TKD AC-220 resistant cells, but not of FLT3-wt cells. Consistently, RNA interference-mediated knock-down of CDC25A reduced the proliferation of FLT3-ITD cell lines. Finally, the clonogenic capacity of primary FLT3-ITD AML cells was reduced by the CDC25 inhibitor IRC-083864, while FLT3-wt AML and normal CD34+ myeloid cells were unaffected. In good agreement, in a cohort of 100 samples from AML patients with intermediate-risk cytogenetics, high levels of CDC25A mRNA were predictive of higher clonogenic potential in FLT3-ITD+ samples, not in FLT3-wt ones.Importantly, pharmacological inhibition as well as RNA interference-mediated knock-down of CDC25A also induced monocytic differentiation of FLT3-ITD positive cells, as judged by cell surface markers expression, morphological modifications, and C/EBPα phosphorylation. CDC25 inhibition also re-induced monocytic differentiation in primary AML blasts carrying the FLT3-ITD mutation, but not in blasts expressing wild type FLT3. Altogether, these data identify CDC25A as an early cell cycle transducer of FLT3-ITD oncogenic signaling, and as a promising target to inhibit proliferation and re-induce differentiation of FLT3-ITD AML cells.
Collapse
Affiliation(s)
- Sarah Bertoli
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Department, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Helena Boutzen
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Laure David
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Clément Larrue
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - François Vergez
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Anne Fernandez-Vidal
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Lingli Yuan
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Marie-Anne Hospital
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - Jérôme Tamburini
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - Cécile Demur
- Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Eric Delabesse
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Estelle Saland
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Jean-Emmanuel Sarry
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | | | - Véronique Mansat-De Mas
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Christine Didier
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Christine Dozier
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Christian Récher
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Department, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Stéphane Manenti
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| |
Collapse
|
20
|
Chen LS, Yang JY, Liang H, Cortes JE, Gandhi V. Protein profiling identifies mTOR pathway modulation and cytostatic effects of Pim kinase inhibitor, AZD1208, in acute myeloid leukemia. Leuk Lymphoma 2016; 57:2863-2873. [PMID: 27054578 DOI: 10.3109/10428194.2016.1166489] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pim kinases phosphorylate and regulate a number of key acute myeloid leukemia (AML) cell survival proteins, and Pim inhibitors have recently entered clinical trial for hematological malignancies. AZD1208 is a small molecule pan-Pim kinase inhibitor and AZD1208 treatment resulted in growth inhibition and cell size reduction in AML cell lines including FLT3-WT (OCI-AML-3, KG-1a, and MOLM-16) and FLT3-ITD mutated (MOLM-13 and MV-4-11). There was limited apoptosis induction (<10% increase) in the AML cell lines evaluated with up to 3 μM AZD1208 for 24 h, suggesting that growth inhibition is not through apoptosis induction. Using reverse phase protein array (RPPA) and immunoblot analysis, we identified that AZD1208 resulted in suppression of mTOR signaling, including inhibition of protein phosphorylation of mTOR (Ser2448), p70S6K (Thr389), S6 (Ser235/236), and 4E-BP1 (Ser65). Consistent with mTOR inhibition, there was also a reduction in protein synthesis that correlated with cell size reduction and growth inhibition with AZD1208; our study provides insights into the mechanism of AZD1208.
Collapse
Affiliation(s)
- Lisa S Chen
- a Department of Experimental Therapeutics , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Ji-Yeon Yang
- b Department of Applied Mathematics , Kumoh National Institute of Technology , Gumi , Korea
| | - Han Liang
- c Department of Bioinformatics and Computational Biology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,d Department of Systems Biology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Jorge E Cortes
- e Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Varsha Gandhi
- a Department of Experimental Therapeutics , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,e Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
21
|
Leung CON, Wong CCL, Fan DNY, Kai AKL, Tung EKK, Xu IMJ, Ng IOL, Lo RCL. PIM1 regulates glycolysis and promotes tumor progression in hepatocellular carcinoma. Oncotarget 2016; 6:10880-92. [PMID: 25834102 PMCID: PMC4484426 DOI: 10.18632/oncotarget.3534] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/18/2015] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is characteristically one of the most rapidly proliferating tumors which outgrows functional blood supply and results in regional oxygen deprivation. Overexpression of PIM1, a serine/threonine kinase, has been identified recently in human cancers. Knowledge on PIM1 in HCC is however, scarce. By immunohistochemical analysis on 56 human primary HCC samples, we observed overexpression of PIM1 in 39% of the cases. In two independent cohorts of paired primary and extra-hepatic metastatic HCC tissues, PIM1 expression was higher (p=0.002) in the extra-hepatic metastatic HCC tissues as compared with the corresponding primary HCCs. PIM1 was markedly up-regulated in multiple HCC cell lines in hypoxic condition (1% O2) versus normoxia (20% O2). Silencing of PIM1 suppressed HCC cell invasion in vitro as compared to non-target control, and decreased HCC cell proliferation in vitro and tumor growth and metastatic potential in vivo. Knockdown of PIM1 significantly reduced glucose uptake by HCC cells and was associated with decreased levels of p-AKT and key molecules in the glycolytic pathway. Taken together, PIM1 is up-regulated by hypoxia in HCC and promotes tumor growth and metastasis through facilitating cancer cell glycolysis. Targeting PIM1 may have potential role in the management of HCC.
Collapse
Affiliation(s)
| | - Carmen Chak-lui Wong
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | | | - Alan Ka-lun Kai
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | | | - Iris Ming-jing Xu
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Irene Oi-lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Regina Cheuk-lam Lo
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
22
|
Kapelko-Slowik K, Owczarek TB, Grzymajlo K, Urbaniak-Kujda D, Jazwiec B, Slowik M, Kuliczkowski K, Ugorski M. Elevated PIM2 gene expression is associated with poor survival of patients with acute myeloid leukemia. Leuk Lymphoma 2016; 57:2140-9. [PMID: 26764044 DOI: 10.3109/10428194.2015.1124991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The PIM2 gene encodes the serine/threonine kinase involved in cell survival and apoptosis. The aim of the study was to evaluate the expression of the PIM2 gene in acute myeloid leukemia (AML) and to examine its role in apoptosis of the blastic cells. We analyzed the PIM2 expression in 148 patients: 91 with AML, 57 with acute lymphoblastic leukemia and 24 healthy controls by Real-Time PCR and Western blot. Inhibition of the PIM2 gene in human leukemic HL60 cell line was performed with RNAi and apoptosis rate was analyzed. Our results indicate that overexpression of PIM2 in AML is associated with low complete remission rate, high-risk cytogenetics, shorter leukemia-free survival, and event-free survival. Cytometric analysis of HL60/PAC-GFP and HL60/PAC-GFP-shPIM2 cells revealed an increase in the number of apoptotic cells after inhibition of PIM2 gene. In summary, the elevated expression of PIM2 in blastic cells is associated with poor prognosis of AML patients and their resistance to induction therapy.
Collapse
Affiliation(s)
- Katarzyna Kapelko-Slowik
- a Department of Hematology, Neoplastic Blood Disorders and Bone Marrow Transplantation , Wroclaw Medical University , Wroclaw , Poland
| | - Tomasz B Owczarek
- b Ludwik Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences , Wroclaw , Poland ;,c Department of Biochemistry, Pharmacology and Toxicology , Wroclaw University of Environmental and Life Sciences , Wroclaw , Poland
| | - Krzysztof Grzymajlo
- c Department of Biochemistry, Pharmacology and Toxicology , Wroclaw University of Environmental and Life Sciences , Wroclaw , Poland
| | - Donata Urbaniak-Kujda
- a Department of Hematology, Neoplastic Blood Disorders and Bone Marrow Transplantation , Wroclaw Medical University , Wroclaw , Poland
| | - Bozena Jazwiec
- a Department of Hematology, Neoplastic Blood Disorders and Bone Marrow Transplantation , Wroclaw Medical University , Wroclaw , Poland
| | - Miroslaw Slowik
- d Department of Ophthalmology , Wroclaw Medical University , Wroclaw , Poland
| | - Kazimierz Kuliczkowski
- a Department of Hematology, Neoplastic Blood Disorders and Bone Marrow Transplantation , Wroclaw Medical University , Wroclaw , Poland
| | - Maciej Ugorski
- b Ludwik Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences , Wroclaw , Poland ;,c Department of Biochemistry, Pharmacology and Toxicology , Wroclaw University of Environmental and Life Sciences , Wroclaw , Poland
| |
Collapse
|
23
|
Control of Pim2 kinase stability and expression in transformed human haematopoietic cells. Biosci Rep 2015; 35:BSR20150217. [PMID: 26500282 PMCID: PMC4672348 DOI: 10.1042/bsr20150217] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/05/2015] [Indexed: 01/02/2023] Open
Abstract
The oncogenic Pim2 kinase is overexpressed in several haematological malignancies, such as multiple myeloma and acute myeloid leukaemia (AML), and constitutes a strong therapeutic target candidate. Like other Pim kinases, Pim2 is constitutively active and is believed to be essentially regulated through its accumulation. We show that in leukaemic cells, the three Pim2 isoforms have dramatically short half-lives although the longer isoform is significantly more stable than the shorter isoforms. All isoforms present a cytoplasmic localization and their degradation was neither modified by broad-spectrum kinase or phosphatase inhibitors such as staurosporine or okadaic acid nor by specific inhibition of several intracellular signalling pathways including Erk, Akt and mTORC1. Pim2 degradation was inhibited by proteasome inhibitors but Pim2 ubiquitination was not detected even by blocking both proteasome activity and protein de-ubiquitinases (DUBs). Moreover, Pyr41, an ubiquitin-activating enzyme (E1) inhibitor, did not stabilize Pim2, strongly suggesting that Pim2 was degraded by the proteasome without ubiquitination. In agreement, we observed that purified 20S proteasome particles could degrade Pim2 molecule in vitro. Pim2 mRNA accumulation in UT7 cells was controlled by erythropoietin (Epo) through STAT5 transcription factors. In contrast, the translation of Pim2 mRNA was not regulated by mTORC1. Overall, our results suggest that Pim2 is only controlled by its mRNA accumulation level. Catalytically active Pim2 accumulated in proteasome inhibitor-treated myeloma cells. We show that Pim2 inhibitors and proteasome inhibitors, such as bortezomib, have additive effects to inhibit the growth of myeloma cells, suggesting that Pim2 could be an interesting target for the treatment of multiple myeloma.
Collapse
|
24
|
Smits VAJ, Gillespie DA. DNA damage control: regulation and functions of checkpoint kinase 1. FEBS J 2015. [DOI: 10.1111/febs.13387] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Veronique A. J. Smits
- Unidad de Investigación; Hospital Universitario de Canarias; Instituto de Tecnologías Biomédicas; Tenerife Spain
| | - David A. Gillespie
- Instituto de Tecnologías Biomédicas; Centro de Investigaciones Biomédicas de Canarias; Facultad de Medicina; Campus Ciencias de la Salud; Universidad de La Laguna; Tenerife Spain
| |
Collapse
|
25
|
Targeting the Pim kinases in multiple myeloma. Blood Cancer J 2015; 5:e325. [PMID: 26186558 PMCID: PMC4526774 DOI: 10.1038/bcj.2015.46] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/11/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy that remains incurable. Novel treatment strategies to improve survival are urgently required. The Pims are a small family of serine/threonine kinases with increased expression across the hematological malignancies. Pim-2 shows highest expression in MM and constitutes a promising therapeutic target. It is upregulated by the bone marrow microenvironment to mediate proliferation and promote MM survival. Pim-2 also has a key role in the bone destruction typically seen in MM. Additional putative roles of the Pim kinases in MM include trafficking of malignant cells, promoting oncogenic signaling in the hypoxic bone marrow microenvironment and mediating resistance to therapy. A number of Pim inhibitors are now under development with lead compounds entering the clinic. The ATP-competitive Pim inhibitor LGH447 has recently been reported to have single agent activity in MM. It is anticipated that Pim inhibition will be of clinical benefit in combination with standard treatments and/or with novel drugs targeting other survival pathways in MM.
Collapse
|
26
|
Esposito MT, So CWE. DNA damage accumulation and repair defects in acute myeloid leukemia: implications for pathogenesis, disease progression, and chemotherapy resistance. Chromosoma 2014; 123:545-61. [PMID: 25112726 DOI: 10.1007/s00412-014-0482-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 12/15/2022]
Abstract
DNA damage repair mechanisms are vital to maintain genomic integrity. Mutations in genes involved in the DNA damage response (DDR) can increase the risk of developing cancer. In recent years, a variety of polymorphisms in DDR genes have been associated with increased risk of developing acute myeloid leukemia (AML) or of disease relapse. Moreover, a growing body of literature has indicated that epigenetic silencing of DDR genes could contribute to the leukemogenic process. In addition, a variety of AML oncogenes have been shown to induce replication and oxidative stress leading to accumulation of DNA damage, which affects the balance between proliferation and differentiation. Conversely, upregulation of DDR genes can provide AML cells with escape mechanisms to the DDR anticancer barrier and induce chemotherapy resistance. The current review summarizes the DDR pathways in the context of AML and describes how aberrant DNA damage response can affect AML pathogenesis, disease progression, and resistance to standard chemotherapy, and how defects in DDR pathways may provide a new avenue for personalized therapeutic strategies in AML.
Collapse
Affiliation(s)
- Maria Teresa Esposito
- Leukemia and Stem Cell Biology Group, Department of Hematological Medicine, King's College London, Denmark Hill campus, SE5 9NU, London, UK
| | | |
Collapse
|
27
|
Yu Z, Zhao X, Ge Y, Zhang T, Huang L, Zhou X, Xie L, Liu J, Huang G. A regulatory feedback loop between HIF-1α and PIM2 in HepG2 cells. PLoS One 2014; 9:e88301. [PMID: 24505470 PMCID: PMC3914973 DOI: 10.1371/journal.pone.0088301] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/06/2014] [Indexed: 12/11/2022] Open
Abstract
To survive under hypoxic conditions, cancer cells remodel glucose metabolism to support tumor progression. HIF transcription factor is essential for cellular response to hypoxia. The underlying mechanism how HIF is constitutively activated in cancer cells remains elusive. In the present study, we characterized a regulatory feedback loop between HIF-1α and PIM2 in HepG2 cells. Serine/threonine kinase proto-oncogene PIM2 level was induced upon hypoxia in a HIF-1α-mediated manner in cancer cells. HIF-1α induced PIM2 expression via binding to the hypoxia-responsive elements (HREs) of the PIM2 promoter. In turn, PIM2 interacted with HIF-1α, especially a transactivation domain of HIF-1α. PIM2 as a co-factor but not an upstream kinase of HIF-1α, enhanced HIF-1α effect in response to hypoxia. The positive feedback loop between PIM2 and HIF-1α was correlated with glucose metabolism as well as cell survival in HepG2 cells. Such a regulatory mode may be important for the adaptive responses of cancer cells in antagonizing hypoxia during cancer progression.
Collapse
Affiliation(s)
- Zhenhai Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoping Zhao
- Department of Nuclear Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Ge
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Teng Zhang
- Department of Nuclear Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liangqian Huang
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Xiang Zhou
- Department of Nuclear Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xie
- Department of Nuclear Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (JL); (GH)
| | - Gang Huang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Department of Nuclear Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
- * E-mail: (JL); (GH)
| |
Collapse
|