1
|
Rathi A, Noor S, Sulaimani MN, Ahmed S, Taiyab A, AlAjmi MF, Khan FI, Hassan MI, Haque MM. FDA-approved drugs as PIM-1 kinase inhibitors: A drug repurposed approach for cancer therapy. Int J Biol Macromol 2024; 292:139107. [PMID: 39722389 DOI: 10.1016/j.ijbiomac.2024.139107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/10/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
PIM-1 kinase, a member of the Serine/Threonine kinase family, has emerged as a promising therapeutic target in various cancers due to its role in promoting tumor growth and resistance to conventional therapies. In this study, we employed a structure-based approach to screen 3800 FDA-approved drugs to discover potential inhibitors of PIM-1. After an initial selection of 50 candidates based on high docking scores, four drugs, stanozolol, alfaxalone, rifaximin, and telmisartan, were identified as strong PIM-1 binders, interacting with key residues in the ATP-binding pocket of the kinase. To assess the stability of these interactions, we conducted all-atom molecular dynamic simulations, confirming favorable dynamics. Experimental validation via a kinase inhibition assay on recombinant PIM-1 showed that rifaximin significantly inhibited PIM-1 activity, with an IC50 of ∼26 μM. Fluorescence binding assays further demonstrated a strong binding affinity for rifaximin, with a binding constant, corroborated by isothermal titration calorimetry studies. Our findings suggest that rifaximin may serve as a potential repurposed drug for targeting PIM-1 in cancer treatment. However, further validations are required in a clinical setting before the final therapeutic implications.
Collapse
Affiliation(s)
- Aanchal Rathi
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Md Nayab Sulaimani
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shahbaz Ahmed
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Mohamed F AlAjmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faez Iqbal Khan
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| | - Mohammad Mahfuzul Haque
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
2
|
Trzaskoma P, Jung S, Pękowska A, Bohrer CH, Wang X, Naz F, Dell’Orso S, Dubois WD, Olivera A, Vartak SV, Zhao Y, Nayak S, Overmiller A, Morasso MI, Sartorelli V, Larson DR, Chow CC, Casellas R, O’Shea JJ. 3D chromatin architecture, BRD4, and Mediator have distinct roles in regulating genome-wide transcriptional bursting and gene network. SCIENCE ADVANCES 2024; 10:eadl4893. [PMID: 39121214 PMCID: PMC11313860 DOI: 10.1126/sciadv.adl4893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 07/08/2024] [Indexed: 08/11/2024]
Abstract
Discontinuous transcription is evolutionarily conserved and a fundamental feature of gene regulation; yet, the exact mechanisms underlying transcriptional bursting are unresolved. Analyses of bursting transcriptome-wide have focused on the role of cis-regulatory elements, but other factors that regulate this process remain elusive. We applied mathematical modeling to single-cell RNA sequencing data to infer bursting dynamics transcriptome-wide under multiple conditions to identify possible molecular mechanisms. We found that Mediator complex subunit 26 (MED26) primarily regulates frequency, MYC regulates burst size, while cohesin and Bromodomain-containing protein 4 (BRD4) can modulate both. Despite comparable effects on RNA levels among these perturbations, acute depletion of MED26 had the most profound impact on the entire gene regulatory network, acting downstream of chromatin spatial architecture and without affecting TATA box-binding protein (TBP) recruitment. These results indicate that later steps in the initiation of transcriptional bursts are primary nodes for integrating gene networks in single cells.
Collapse
Affiliation(s)
- Pawel Trzaskoma
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - SeolKyoung Jung
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aleksandra Pękowska
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Dioscuri Centre for Chromatin Biology and Epigenomics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | | | - Xiang Wang
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Faiza Naz
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stefania Dell’Orso
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wendy D. Dubois
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ana Olivera
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Supriya V. Vartak
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yongbing Zhao
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Subhashree Nayak
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Overmiller
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria I. Morasso
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vittorio Sartorelli
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel R. Larson
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carson C. Chow
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rafael Casellas
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John J. O’Shea
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
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
|
4
|
Karati D, Saha A, Roy S, Mukherjee S. PIM Kinase Inhibitors as Novel Promising Therapeutic Scaffolds in Cancer Therapy. Curr Top Med Chem 2024; 24:2489-2508. [PMID: 39297470 DOI: 10.2174/0115680266321659240906114742] [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: 04/06/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 11/21/2024]
Abstract
Cancer involves the uncontrolled, abnormal growth of cells and affects other tissues. Kinase has an impact on proliferating the cells and causing cancer. For the purpose of treating cancer, PIM kinase is a potential target. The pro-viral Integration site for moloney murine leukaemia virus (PIM) kinases is responsible for the tumorigenesis, by phosphorylating the proteins that control the cell cycle and cell proliferation. PIM-1, PIM-2, and PIM-3 are the three distinct isoforms of PIM kinases. The JAK/STAT pathway is essential for controlling how PIM genes are expressed. PIM kinase is also linked withPI3K/AKT/mTOR pathway in various types of cancers. The overexpression of PIM kinase will cause cancer. Currently, there are significant efforts being made in medication design and development to target its inhibition. A few small chemical inhibitors (E.g., SGI-1776, AZD1208, LGH447) that specifically target the PIM proteins' adenosine triphosphate (ATP)-binding domain have been identified. PIM kinase antagonists have a remarkable effect on different types of cancer. Despite conducting clinical trials on SGI-1776, the first PIM inhibitory agent, was prematurely withdrawn, making it unable to generate concept evidence. On the other hand, in recent years, it has aided in hastening the identification of multiple new PIM inhibitors. Cyanopyridines and Pyrazolo[1,5-a]pyrimidinecan act as potent PIM kinase inhibitors for cancer therapy. We explore the involvement of oncogenic transcription factor c-Mycandmi-RNA in relation to PIM kinase. In this article, we highlight the oncogenic effects, and structural insights into PIM kinase inhibitors for the treatment of cancer.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata 700091, West Bengal, India
| | - Ankur Saha
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata 700091, West Bengal, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata 700053, West Bengal, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata 700053, West Bengal, India
| |
Collapse
|
5
|
Nock S, Karim E, Unsworth AJ. Pim Kinases: Important Regulators of Cardiovascular Disease. Int J Mol Sci 2023; 24:11582. [PMID: 37511341 PMCID: PMC10380471 DOI: 10.3390/ijms241411582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Pim Kinases; Pim-1, Pim-2, and Pim-3, are a family of constitutively active serine/threonine kinases, widely associated with cell survival, proliferation, and migration. Historically considered to be functionally redundant, independent roles for the individual isoforms have been described. Whilst most established for their role in cancer progression, there is increasing evidence for wider pathological roles of Pim kinases within the context of cardiovascular disease, including inflammation, thrombosis, and cardiac injury. The Pim kinase isoforms have widespread expression in cardiovascular tissues, including the heart, coronary artery, aorta, and blood, and have been demonstrated to be upregulated in several co-morbidities/risk factors for cardiovascular disease. Pim kinase inhibition may thus be a desirable therapeutic for a multi-targeted approach to treat cardiovascular disease and some of the associated risk factors. In this review, we discuss what is known about Pim kinase expression and activity in cells of the cardiovascular system, identify areas where the role of Pim kinase has yet to be fully explored and characterised and review the suitability of targeting Pim kinase for the prevention and treatment of cardiovascular events in high-risk individuals.
Collapse
Affiliation(s)
| | | | - Amanda J. Unsworth
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
| |
Collapse
|
6
|
Castanet AS, Nafie MS, Said SA, Arafa RK. Discovery of PIM-1 kinase inhibitors based on the 2,5-disubstituted 1,3,4-oxadiazole scaffold against prostate cancer: Design, synthesis, in vitro and in vivo cytotoxicity investigation. Eur J Med Chem 2023; 250:115220. [PMID: 36848846 DOI: 10.1016/j.ejmech.2023.115220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
PIM-1 kinases play an established role in prostate cancer development and progression. This research work tackles the design and synthesis of new PIM-1 kinase targeting 2,5-disubstituted-1,3,4-oxadiazoles 10a-g&11a-f, and investigation thereof as potential anti-cancer agents through in vitro cytotoxicity assay followed by in vivo studies along with exploration of this chemotype's plausible mechanism of action. In vitro cytotoxicity experiments have disclosed 10f as the most potent derivative against PC-3 cells (IC50 = 16 nM) compared to the reference drug Staurosporine (IC50 = 0.36 μM), also eliciting good cytotoxicity against HepG2 and MCF-7 cells (IC50 = 0.13 and 5.37 μM, respectively). Investigating PIM-1 kinase inhibitory activity of compound 10f revealed an IC50 of 17 nM paralleled to that of Staurosporine (IC50 = 16.7 nM). Furthermore, compound 10f displayed an antioxidant activity eliciting a DPPH inhibition ratio of 94% as compared to Trolox (96%). Further investigation demonstrated that 10f induced apoptosis in treated PC-3 cells by 43.2-fold (19.44%) compared to 0.45% in control. 10f also disrupted the PC-3 cell cycle by increasing the cell population at the PreG1-phase by 19.29-fold while decreasing the G2/M-phase by 0.56-fold compared to control. Moreover, 10f affected a downregulation of JAK2, STAT3 and Bcl-2 and upregulation of caspases 3, 8 and 9 levels that activated the caspase-dependent apoptosis. Finally, in vivo 10f-treatment caused a significant increase in tumor inhibition by 64.2% compared to 44.5% in Staurosporine treatment of the PC-3 xenograft mouse model. Additionally, it improved the hematological, biochemical parameters, and histopathological examinations compared to control untreated animals. Finally, docking of 10f with the ATP-binding site of PIM-1 kinase demonstrated good recognition of and effective binding to the active site. In conclusion, compound 10f represents a promising lead compound that merits further future optimization for controlling prostate cancer.
Collapse
Affiliation(s)
- Anne-Sophie Castanet
- Institut des Molécules et Matériaux du Mans, IMMM-UMR 6283 CNRS, Le Mans Université, Avenue Olivier Messiaen, 72085, LE MANS CEDEX 9, France
| | - Mohamed S Nafie
- Chemistry Department (Biochemistry program), Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| | - Sara A Said
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Reem K Arafa
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Giza, 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
| |
Collapse
|
7
|
New Quinoxaline Derivatives as Dual Pim-1/2 Kinase Inhibitors: Design, Synthesis and Biological Evaluation. Molecules 2021; 26:molecules26040867. [PMID: 33562106 PMCID: PMC7914722 DOI: 10.3390/molecules26040867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 11/17/2022] Open
Abstract
Proviral integration site for Moloney murine leukemia virus (Pim)-1/2 kinase overexpression has been identified in a variety of hematologic (e.g., multiple myeloma or acute myeloid leukemia (AML)) and solid (e.g., colorectal carcinoma) tumors, playing a key role in cancer progression, metastasis, and drug resistance, and is linked to poor prognosis. These kinases are thus considered interesting targets in oncology. We report herein the design, synthesis, structure–activity relationships (SAR) and in vitro evaluations of new quinoxaline derivatives, acting as dual Pim1/2 inhibitors. Two lead compounds (5c and 5e) were then identified, as potent submicromolar Pim-1 and Pim-2 inhibitors. These molecules were also able to inhibit the growth of the two human cell lines, MV4-11 (AML) and HCT-116 (colorectal carcinoma), expressing high endogenous levels of Pim-1/2 kinases.
Collapse
|
8
|
Sugita M, Onishi I, Irisa M, Yoshida N, Hirata F. Molecular Recognition and Self-Organization in Life Phenomena Studied by a Statistical Mechanics of Molecular Liquids, the RISM/3D-RISM Theory. Molecules 2021; 26:E271. [PMID: 33430461 PMCID: PMC7826681 DOI: 10.3390/molecules26020271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 11/18/2022] Open
Abstract
There are two molecular processes that are essential for living bodies to maintain their life: the molecular recognition, and the self-organization or self-assembly. Binding of a substrate by an enzyme is an example of the molecular recognition, while the protein folding is a good example of the self-organization process. The two processes are further governed by the other two physicochemical processes: solvation and the structural fluctuation. In the present article, the studies concerning the two molecular processes carried out by Hirata and his coworkers, based on the statistical mechanics of molecular liquids or the RISM/3D-RISM theory, are reviewed.
Collapse
Affiliation(s)
- Masatake Sugita
- Department of Computer Science, School of Computing, Tokyo Institute of Technology, W8-76, 2-12-1, Ookayama Meguro-ku, Tokyo 152-8550, Japan;
| | - Itaru Onishi
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan; (I.O.); (M.I.)
| | - Masayuki Irisa
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan; (I.O.); (M.I.)
| | - Norio Yoshida
- Department of Chemistry, Kyushu University, Fukuoka, Fukuoka 812-8581, Japan;
| | - Fumio Hirata
- Theoretical and Computational Molecular Science, Institute for Molecular Science, Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
9
|
Alnabulsi S, Al-Hurani EA. Pim kinase inhibitors in cancer: medicinal chemistry insights into their activity and selectivity. Drug Discov Today 2020; 25:S1359-6446(20)30374-3. [PMID: 32971234 DOI: 10.1016/j.drudis.2020.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/09/2020] [Accepted: 09/14/2020] [Indexed: 01/09/2023]
Abstract
The oncogenic Pim kinase proteins (Pim-1/2/3) regulate tumorigenesis through phosphorylating essential proteins that control cell cycle and proliferation. Pim kinase is a potential chemotherapeutic target in cancer and its inhibition is currently the focus of intensive drug design and development efforts. The distinctive presence of proline amino acids in the hinge region provides an opportunity to inhibit Pim kinase while conserving the physiological functions of other kinases and reducing the toxicity profiles of the inhibitors. Various Pim kinase inhibitors have been clinically evaluated for the treatment of hematological cancers, yet none has reached the clinic. In this review, we discuss the design and development of selective and potent Pim inhibitors with novel chemotypes focusing on structural features essential for high potency and selectivity.
Collapse
Affiliation(s)
- Soraya Alnabulsi
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan.
| | - Enas A Al-Hurani
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| |
Collapse
|
10
|
Kim S, Kim W, Kim DH, Jang JH, Kim SJ, Park SA, Hahn H, Han BW, Na HK, Chun KS, Choi BY, Surh YJ. Resveratrol suppresses gastric cancer cell proliferation and survival through inhibition of PIM-1 kinase activity. Arch Biochem Biophys 2020; 689:108413. [PMID: 32473133 DOI: 10.1016/j.abb.2020.108413] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/01/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
The proviral integration site for Moloney murine leukemia virus (PIM) family of serine/threonine-specific kinases consist of three isoforms, that regulate proliferation, apoptosis, metabolism, invasion, and metastasis of cancer cells. Among these, abnormally elevated kinase activity of PIM-1 contributes to the progression of gastric cancer and predicts poor prognosis and a low survival rate in gastric cancer patients. In the present study, we found that resveratrol, one of the representative chemopreventive and anticarcinogenic phytochemicals, directly binds to PIM-1 and thereby inhibits its catalytic activity in human gastric cancer SNU-601 cells. This resulted in suppression of phosphorylation of the proapoptotic Bad, a known substrate of PIM-1. Resveratrol, by inactivating PIM-1, also inhibited anchorage-independent growth and proliferation of SNU-601 cells. To understand the molecular interaction between resveratrol and PIM-1, we conducted docking simulation and found that resveratrol directly binds to the PIM-1 at the ATP-binding pocket. In conclusion, the proapototic and anti-proliferative effects of resveratrol in gastric cancer cells are likely to be mediated through suppression of PIM-1 kinase activity, which may represent a novel mechanism underlying its chemopreventive and anticarcinogenic actions.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Wonki Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do 16227, South Korea
| | - Jeong-Hoon Jang
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Sin-Aye Park
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, South Korea
| | - Hyunggu Hahn
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Byung Woo Han
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-based Services Engineering, Sungshin Women's University, Seoul 01133, South Korea
| | - Kyung-Soo Chun
- Department of Pharmacy, College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| | - Bu Young Choi
- Department of Pharmaceutical Science and Engineering, Seowon University, Cheongju, Chungbuk 28674, South Korea.
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Cancer Research Institute, Seoul National University, Seoul 03080, South Korea.
| |
Collapse
|
11
|
Ashida T, Kikuchi T. A new method for estimating the relative binding free energy, derived from a free energy variational principle for the Pim-1-kinase-ligand and FKBP-ligand systems. J Comput Aided Mol Des 2020; 34:647-658. [PMID: 32107701 DOI: 10.1007/s10822-020-00302-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/14/2020] [Indexed: 11/26/2022]
Abstract
In this study, a new method is proposed for calculating the relative binding free energy between a ligand and a protein, derived from a free energy variational principle (FEVP). To address the shortcomings of the method used in our previous study, we incorporate the dynamical fluctuation of a ligand in the FEVP calculation. The present modified method is applied to the Pim-1-kinase-ligand system and also to the FKBP-ligand system as a comparison with our previous work. Any inhibitor of Pim-1 kinase is expected to function as an anti-cancer drug. Some improvements are observed in the results compared to the previous study. The present work also shows comparable or better results than approaches using a standard technique of binding free energy calculations, such as the LIE and the MM-PB/SA methods. The possibility of applying the present method in the drug discovery process is also discussed.
Collapse
Affiliation(s)
- Takeshi Ashida
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Takeshi Kikuchi
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
12
|
Liang C, Shi S, Meng Q, Liang D, Hua J, Qin Y, Zhang B, Xu J, Ni Q, Yu X. MiR‐29a, targeting caveolin 2 expression, is responsible for limitation of pancreatic cancer metastasis in patients with normal level of serum CA125. Int J Cancer 2018; 143:2919-2931. [DOI: 10.1002/ijc.31654] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/29/2018] [Indexed: 08/29/2023]
Affiliation(s)
- Chen Liang
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Si Shi
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Qingcai Meng
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Dingkong Liang
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Jie Hua
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Yi Qin
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Bo Zhang
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Jin Xu
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Quanxing Ni
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| | - Xianjun Yu
- Department of Pancreatic Surgery Fudan University Shanghai Cancer Center Shanghai 200032 China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai 200032 China
- Shanghai Pancreatic Cancer Institute Shanghai 200032 China
- Pancreatic Cancer Institute Fudan University Shanghai 200032 China
| |
Collapse
|
13
|
Carlson DA, Singer MR, Sutherland C, Redondo C, Alexander LT, Hughes PF, Knapp S, Gurley SB, Sparks MA, MacDonald JA, Haystead TAJ. Targeting Pim Kinases and DAPK3 to Control Hypertension. Cell Chem Biol 2018; 25:1195-1207.e32. [PMID: 30033129 PMCID: PMC6863095 DOI: 10.1016/j.chembiol.2018.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 03/16/2018] [Accepted: 06/20/2018] [Indexed: 01/19/2023]
Abstract
Sustained vascular smooth muscle hypercontractility promotes hypertension and cardiovascular disease. The etiology of hypercontractility is not completely understood. New therapeutic targets remain vitally important for drug discovery. Here we report that Pim kinases, in combination with DAPK3, regulate contractility and control hypertension. Using a co-crystal structure of lead molecule (HS38) in complex with DAPK3, a dual Pim/DAPK3 inhibitor (HS56) and selective DAPK3 inhibitors (HS94 and HS148) were developed to provide mechanistic insight into the polypharmacology of hypertension. In vitro and ex vivo studies indicated that Pim kinases directly phosphorylate smooth muscle targets and that Pim/DAPK3 inhibition, unlike selective DAPK3 inhibition, significantly reduces contractility. In vivo, HS56 decreased blood pressure in spontaneously hypertensive mice in a dose-dependent manner without affecting heart rate. These findings suggest including Pim kinase inhibition within a multi-target engagement strategy for hypertension management. HS56 represents a significant step in the development of molecularly targeted antihypertensive medications.
Collapse
Affiliation(s)
- David A Carlson
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Miriam R Singer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Cindy Sutherland
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Clara Redondo
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK
| | - Leila T Alexander
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stefan Knapp
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK; Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - Susan B Gurley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC 27710, USA
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC 27710, USA
| | - Justin A MacDonald
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
14
|
Oyallon B, Brachet-Botineau M, Logé C, Bonnet P, Souab M, Robert T, Ruchaud S, Bach S, Berthelot P, Gouilleux F, Viaud-Massuard MC, Denevault-Sabourin C. Structure-based design of novel quinoxaline-2-carboxylic acids and analogues as Pim-1 inhibitors. Eur J Med Chem 2018; 154:101-109. [PMID: 29778892 DOI: 10.1016/j.ejmech.2018.04.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/20/2018] [Accepted: 04/28/2018] [Indexed: 12/28/2022]
Abstract
We identified a new series of quinoxaline-2-carboxylic acid derivatives, targeting the human proviral integration site for Moloney murine leukemia virus-1 (HsPim-1) kinase. Seventeen analogues were synthesized providing useful insight into structure-activity relationships studied. Docking studies realized in the ATP pocket of HsPim-1 are consistent with an unclassical binding mode of these inhibitors. The lead compound 1 was able to block HsPim-1 enzymatic activity at nanomolar concentrations (IC50 of 74 nM), with a good selectivity profile against a panel of mammalian protein kinases. In vitro studies on the human chronic myeloid leukemia cell line KU812 showed an antitumor activity at micromolar concentrations. As a result, compound 1 represents a promising lead for the design of novel anticancer targeted therapies.
Collapse
Affiliation(s)
- Bruno Oyallon
- EA GICC - ERL 7001 CNRS « Groupe Innovation et Ciblage Cellulaire », Team Innovation Moléculaire et Thérapeutique, University of Tours, F-37200, Tours, France
| | - Marie Brachet-Botineau
- CNRS ERL7001 LNOx « Leukemic Niche and RedOx Metabolism » - EA GICC, University of Tours, F-37000, Tours, France; CHRU de Tours, Service d'Hématologie Biologique, F-37044, Tours, France
| | - Cédric Logé
- Université de Nantes, Nantes Atlantique Universités, Département de Chimie Thérapeutique, Cibles et Médicaments des Infections et du Cancer, IICIMED- EA1155, Institut de Recherche en Santé 2, F-44200, Nantes, France
| | - Pascal Bonnet
- UMR University of Orléans-CNRS 7311, Institut de Chimie Organique et Analytique (ICOA), University of Orléans, F-45067, Orléans, France
| | - Mohamed Souab
- Sorbonne Universités, USR3151 CNRS/UPMC, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique, Place Georges Teissier, F-29688, Roscoff, France
| | - Thomas Robert
- Sorbonne Universités, USR3151 CNRS/UPMC, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique, Place Georges Teissier, F-29688, Roscoff, France
| | - Sandrine Ruchaud
- Sorbonne Universités, USR3151 CNRS/UPMC, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique, Place Georges Teissier, F-29688, Roscoff, France
| | - Stéphane Bach
- Sorbonne Universités, USR3151 CNRS/UPMC, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique, Place Georges Teissier, F-29688, Roscoff, France
| | - Pascal Berthelot
- UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, University of Lille, Inserm, CHU Lille, F-59000, Lille, France
| | - Fabrice Gouilleux
- CNRS ERL7001 LNOx « Leukemic Niche and RedOx Metabolism » - EA GICC, University of Tours, F-37000, Tours, France
| | - Marie-Claude Viaud-Massuard
- EA GICC - ERL 7001 CNRS « Groupe Innovation et Ciblage Cellulaire », Team Innovation Moléculaire et Thérapeutique, University of Tours, F-37200, Tours, France
| | - Caroline Denevault-Sabourin
- EA GICC - ERL 7001 CNRS « Groupe Innovation et Ciblage Cellulaire », Team Innovation Moléculaire et Thérapeutique, University of Tours, F-37200, Tours, France.
| |
Collapse
|
15
|
Wilson JRF, Saeed F, Tyagi AK, Goodden JR, Sivakumar G, Crimmins D, Elliott M, Picton S, Chumas PD. Pre-operative neutrophil count and neutrophil-lymphocyte count ratio (NLCR) in predicting the histological grade of paediatric brain tumours: a preliminary study. Acta Neurochir (Wien) 2018; 160:793-800. [PMID: 29188366 PMCID: PMC5859055 DOI: 10.1007/s00701-017-3388-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/31/2017] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The neutrophil-lymphocyte count ratio (NLCR) is an established prognostic marker for renal, lung and colorectal carcinomas and has been suggested to be predictive of histological grade and outcome in adult intracranial tumours. The purpose of this study was to determine whether a correlation of the pre-operative neutrophil count (NC) and NLCR with the final histological grade exists in paediatric intracranial tumours. METHODS A retrospective analysis was undertaken at a single centre. Patients less than 18 years old at the time of surgery who underwent tumour-related procedures from 2006 to 2015 were included. Patients with recurrent tumours, previous bone marrow transplant and metastases were excluded. Pre-operative full blood counts (FBC), collected before the diagnosis of intracranial pathology and before administration of steroids, were matched with histological diagnosis for each patient. Post-operative FBC was also recorded, together with survival data where applicable. RESULTS A total of 116 patients (74 male, 42 female; mean age, 8 ± 0.9 years) with a diagnosis of primary intracranial tumours had pre-operative FBC that could be matched to final histological grade. Pre-operative NC and NLCR were higher with increasing grade of tumour: grade 1 (NC 4.29 109/l, NLCR 2.26), grade 2 (NC 4.59 109/l, NLCR 2.38), grade 3 (NC 5.67 109/l, NLCR 2.72) and grade 4 (NC 6.59 109/l, NLCR 3.31). Patients with WHO grade 1 and 2 tumours pooled together had a lower NC (4.37 95% CI ± 0.67 109/l) compared to WHO grade 3 and 4 patients (6.41 95% CI ± 0.99 109/l, p = 0.0013). The NLCR was lower in grade 1 and 2 tumours (2.29 ± 0.59) (compared to grade 3 and 4 tumours; 3.20 ± 0.76) but this did not reach significance (p = 0.069). The subgroup of patients with pilocytic astrocytoma had a significantly lower NC when compared to patients with high-grade tumours (p = 0.005). Medulloblastoma and supratentorial PNET subgroups had significantly higher NC compared to the low-grade group (p = 0.033, p = 0.002). Post-operative NC was significantly higher in the high-grade tumours (p = 0.034), but no difference was observed for NLCR (p = 0.28). CONCLUSIONS No evidence exists to support the correlation of pre-operative NC or NLCR to histological diagnosis in paediatric intracranial tumours. Our results indicate that a higher pre-operative NC/NLCR correlates with a higher histological grade of tumour. This suggests that immunological mechanisms may be involved in the pathogenesis of paediatric brain tumours, and a further prospective study is required to substantiate and expand these findings.
Collapse
Affiliation(s)
- J R F Wilson
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, LS13EX, UK.
| | - F Saeed
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, LS13EX, UK
| | - A K Tyagi
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, LS13EX, UK
| | - J R Goodden
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, LS13EX, UK
| | - G Sivakumar
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, LS13EX, UK
| | - D Crimmins
- Department of Neurosurgery, Beaumont Hospital, Dublin, Ireland
| | - M Elliott
- Department of Paediatric Oncology and Haematology, Leeds Children's Hospital, Leeds General Infirmary, Leeds, LS13EX, UK
| | - S Picton
- Department of Paediatric Oncology and Haematology, Leeds Children's Hospital, Leeds General Infirmary, Leeds, LS13EX, UK
| | - P D Chumas
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, LS13EX, UK
| |
Collapse
|
16
|
Xu J, Zhu X, Li Q, Chen C, Guo Z, Tan Z, Zheng C, Ge M. Loss of PIM1 correlates with progression and prognosis of salivary adenoid cystic carcinoma (SACC). Cancer Cell Int 2018; 18:22. [PMID: 29467592 PMCID: PMC5819291 DOI: 10.1186/s12935-018-0518-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/03/2018] [Indexed: 12/31/2022] Open
Abstract
Background Increasing evidence indicates that PIM1 is a potential prognostic marker and target for cancer treatment but its precise mechanisms of action remain to be determined in salivary adenoid cystic carcinoma (SACC). This study aims to decipher the prognostic and mechanistic role of PIM1 in progression of SACC cells and tumor tissues. Methods A SACC cell line (ACC-M) was transfected with shRNA plasmids targeting the PIM1 gene. The expression levels of PIM1, RUNX3 and p21 were measured by quantitative real-time PCR and western blot. Subcellular translocalization of RUNX3 and p21 proteins was assessed using immunofluorescence, and cell cycle phase was quantified using flow cytometry. A total of 97 SACC patients were retrospectively analyzed by clinicopathologic characteristics and survival outcomes. Results After down-regulation of PIM1 in ACC-M cells, RUNX3 and p21 proteins were translocated from cytoplasm to nucleus, with a decrease of p21 expression and increase of G0/G1 phase cells. PIM1 and RUNX3 levels show a distinct covariance. PIM1 is associated with T-status, lymph node involvement, nerve invasion, and distant metastasis in SACC tissues. Patients with low PIM1 level had a better outcome than those with higher PIM1 level. Conclusions PIM1 is multifunctional in ACC-M cells and it serves as a neoteric therapeutic target and potential prognostic marker for SACC patients.
Collapse
Affiliation(s)
- Jiajie Xu
- 1Department of Head and Neck Surgery, Zhejiang Cancer Hospital, No. 38 Guangji Road, Hangzhou, 310022 Zhejiang China
| | - Xin Zhu
- 2Zhejiang Cancer Research Institute, Hangzhou, 310022 China
| | - Qingling Li
- 3Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022 China
| | - Chao Chen
- 1Department of Head and Neck Surgery, Zhejiang Cancer Hospital, No. 38 Guangji Road, Hangzhou, 310022 Zhejiang China
| | - Zhenying Guo
- 4Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022 China
| | - Zhuo Tan
- 1Department of Head and Neck Surgery, Zhejiang Cancer Hospital, No. 38 Guangji Road, Hangzhou, 310022 Zhejiang China
| | - Chuanming Zheng
- 1Department of Head and Neck Surgery, Zhejiang Cancer Hospital, No. 38 Guangji Road, Hangzhou, 310022 Zhejiang China
| | - Minghua Ge
- 1Department of Head and Neck Surgery, Zhejiang Cancer Hospital, No. 38 Guangji Road, Hangzhou, 310022 Zhejiang China
| |
Collapse
|
17
|
Hasegawa T, Sugita M, Kikuchi T, Hirata F. A Systematic Analysis of the Binding Affinity between the Pim-1 Kinase and Its Inhibitors Based on the MM/3D-RISM/KH Method. J Chem Inf Model 2017; 57:2789-2798. [PMID: 29019402 DOI: 10.1021/acs.jcim.7b00158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A systematic study of the binding affinities of 16 lead compounds targeting the Pim-1 kinase based on the 3D-RISM/KH theory and MD simulations is reported. The results show a correlation coefficient R = 0.69 between the theoretical and experimental values of the binding free energy. This demonstrates that the method is applicable to the problem of compound screening and lead optimization, for which relative values of the free energy among the compounds have significance. We elucidate the contribution of the ligand fragments to the binding free energy. Our results indicate that the interactions between the residues and the triazolo[4,3-b]pyridazine scaffold as well as the phenyl ring of the ligand molecule make significant contributions to stabilization of the complex. Using the 3D-RISM/KH theory, we further analyze the probability distribution of a ligand fragment around the protein-ligand complex in which the substituent around the phenyl ring is removed from the ligand. The results demonstrate that the 3D-RISM/KH theory is capable of predicting the position of substitution on a ligand that has a higher affinity to a target protein.
Collapse
Affiliation(s)
- Takeshi Hasegawa
- Department of Bioinformatics, College of Life Science, Ritsumeikan University , 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Masatake Sugita
- Department of Bioinformatics, College of Life Science, Ritsumeikan University , 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Takeshi Kikuchi
- Department of Bioinformatics, College of Life Science, Ritsumeikan University , 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Fumio Hirata
- Toyota Physical and Chemical Research Institute , 41-1 Yokomichi, Nagakute, Aichi 480-1192, Japan
| |
Collapse
|
18
|
Jiménez-García MP, Lucena-Cacace A, Robles-Frías MJ, Narlik-Grassow M, Blanco-Aparicio C, Carnero A. The role of PIM1/PIM2 kinases in tumors of the male reproductive system. Sci Rep 2016; 6:38079. [PMID: 27901106 PMCID: PMC5128923 DOI: 10.1038/srep38079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/03/2016] [Indexed: 12/18/2022] Open
Abstract
The PIM family of serine/threonine kinases has three highly conserved isoforms (PIM1, PIM2 and PIM3). PIM proteins are regulated through transcription and stability by JAK/STAT pathways and are overexpressed in hematological malignancies and solid tumors. The PIM kinases possess weak oncogenic abilities, but enhance other genes or chemical carcinogens to induce tumors. We generated conditional transgenic mice that overexpress PIM1 or PIM2 in male reproductive organs and analyzed their contribution to tumorigenesis. We found an increase in alterations of sexual organs and hyperplasia in the transgenic mice correlating with inflammation. We also found that PIM1/2 are overexpressed in a subset of human male germ cells and prostate tumors correlating with inflammatory features and stem cell markers. Our data suggest that PIM1/2 kinase overexpression is a common feature of male reproductive organs tumors, which provoke tissue alterations and a large inflammatory response that may act synergistically during the process of tumorigenesis. There is also a correlation with markers of cancer stem cells, which may contribute to the therapy resistance found in tumors overexpressing PIM kinases.
Collapse
Affiliation(s)
- Manuel Pedro Jiménez-García
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - Antonio Lucena-Cacace
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - María José Robles-Frías
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - Maja Narlik-Grassow
- Experimental Therapeutics Programme, Spanish National Cancer Centre (CNIO), C/Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Centre (CNIO), C/Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| |
Collapse
|
19
|
Liang C, Yu XJ, Guo XZ, Sun MH, Wang Z, Song Y, Ni QX, Li HY, Mukaida N, Li YY. MicroRNA-33a-mediated downregulation of Pim-3 kinase expression renders human pancreatic cancer cells sensitivity to gemcitabine. Oncotarget 2016; 6:14440-55. [PMID: 25971209 PMCID: PMC4546478 DOI: 10.18632/oncotarget.3885] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/25/2015] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers, with less than 5% of patients surviving 5 years beyond diagnosis. Systemic therapies, particularly gemcitabine, have a modest clinical benefit, but chemoresistance limits their efficacy. Here, we demonstrate that plasma miR-33a levels positively correlated with miR-33a levels in tumor tissues of patients with PDAC and are a good prognostic indicator of overall survival. Overexpression of miR-33a inhibited tumor cell proliferation and increased the chemosensitivity to gemcitabine both in vitro and in vivo. Moreover, miR-33a targets Pim-3 directly in PDAC. Pim-3 expression was a prognostic indicator related to poor survival in pancreatic cancer patients. Plasma miR-33a levels were significantly lower in pancreatic cancer patients with high Pim-3 protein expression than in healthy controls. Furthermore, overexpression of miR-33a in pancreatic cancer cell lines suppressed Pim-3 expression, leading to downregulation of the AKT/Gsk-3β/β-catenin pathway. Overall, these results indicate that miR-33a functions as a tumor suppressor that downregulates Pim-3 kinase expression to inhibit both pancreatic tumor growth and gemcitabine resistance via the AKT/β-catenin pathway. Hence, detection of plasma miR-33a may be a simple and convenient method of predicting therapeutic responses.
Collapse
Affiliation(s)
- Chen Liang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xian-Jun Yu
- Department of Pancreas and Hepatobiliary, Pancreatic Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Zhong Guo
- Department of Gastroenterology, General Hospital of Shenyang Military Area, Shenyang, Liaoning, China
| | - Meng-Hong Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Song
- Division of Molecular Bioregulation, Cancer Microenvironment Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Quan-Xing Ni
- Department of Pancreas and Hepatobiliary, Pancreatic Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Yu Li
- Department of Gastroenterology, General Hospital of Shenyang Military Area, Shenyang, Liaoning, China
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Microenvironment Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Ying-Yi Li
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Division of Molecular Bioregulation, Cancer Microenvironment Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
20
|
Tursynbay Y, Zhang J, Li Z, Tokay T, Zhumadilov Z, Wu D, Xie Y. Pim-1 kinase as cancer drug target: An update. Biomed Rep 2015; 4:140-146. [PMID: 26893828 DOI: 10.3892/br.2015.561] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022] Open
Abstract
Proviral integration site for Moloney murine leukemia virus-1 (Pim-1) is a serine/threonine kinase that regulates multiple cellular functions such as cell cycle, cell survival, drug resistance. Aberrant elevation of Pim-1 kinase is associated with numerous types of cancer. Two distinct isoforms of Pim-1 (Pim-1S and Pim-1L) show distinct cellular functions. Pim-1S predominately localizes to the nucleus and Pim-1L localizes to plasma membrane for drug resistance. Recent studies show that mitochondrial Pim-1 maintains mitochondrial integrity. Pim-1 is emerging as a cancer drug target, particularly in prostate cancer. Recently the potent new functions of Pim-1 in immunotherapy, senescence bypass, metastasis and epigenetic dynamics have been found. The aim of the present updated review is to provide brief information regarding networks of Pim-1 kinase and focus on its recent advances as a novel drug target.
Collapse
Affiliation(s)
- Yernar Tursynbay
- Department of Biology, Nazarbayev University School of Science and Technology, Astana 010000, Republic of Kazakhstan
| | - Jinfu Zhang
- Institute of International Medical Research, Department of Urology and Andrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Zhi Li
- Department of Pathology, Sun Yat-sen University, Guangzhou 510080, P.R. China
| | - Tursonjan Tokay
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Republic of Kazakhstan
| | - Zhaxybay Zhumadilov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Republic of Kazakhstan
| | - Denglong Wu
- Department of Urology, Tong Ji Hospital, Tong Ji University, Shanghai 200065, P.R. China
| | - Yingqiu Xie
- Department of Biology, Nazarbayev University School of Science and Technology, Astana 010000, Republic of Kazakhstan
| |
Collapse
|
21
|
Liang C, Wang Z, Li YY, Yu BH, Zhang F, Li HY. miR-33a suppresses the nuclear translocation of β-catenin to enhance gemcitabine sensitivity in human pancreatic cancer cells. Tumour Biol 2015; 36:9395-403. [DOI: 10.1007/s13277-015-3679-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/15/2015] [Indexed: 01/07/2023] Open
|