1
|
Meur S, Mukherjee S, Roy S, Karati D. Role of PIM Kinase Inhibitor in the Treatment of Alzheimer's Disease. Mol Neurobiol 2024; 61:10941-10955. [PMID: 38816674 DOI: 10.1007/s12035-024-04257-7] [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: 01/08/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, is the most prevalent form of senile dementia, causing progressive deterioration of cognition, behavior, and rational skills. Neuropathologically, AD is characterized by two hallmark proteinaceous aggregates: amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) formed of hyperphosphorylated tau. A significant study has been done to understand how Aβ and/or tau accumulation can alter signaling pathways that affect neuronal function. A conserved protein kinase known as the mammalian target of rapamycin (mTOR) is essential for maintaining the proper balance between protein synthesis and degradation. Overwhelming evidence shows mTOR signaling's primary role in age-dependent cognitive decline and the pathogenesis of AD. Postmortem human AD brains consistently show an upregulation of mTOR signaling. Confocal microscopy findings demonstrated a direct connection between mTOR and intraneuronal Aβ42 through molecular processes of PRAS40 phosphorylation. By attaching to the mTORC1 complex, PRAS40 inhibits the activity of mTOR. Furthermore, inhibiting PRAS40 phosphorylation can stop the Aβ-mediated increase in mTOR activity, indicating that the accumulation of Aβ may aid in PRAS40 phosphorylation. Physiologically, PRAS40 is phosphorylated by PIM1 which is a serine/threonine kinase of proto-oncogene PIM kinase family. Pharmacological inhibition of PIM1 activity prevents the Aβ-induced mTOR hyperactivity in vivo by blocking PRAS40 phosphorylation and restores cognitive impairments by enhancing proteasome function. Recently identified small-molecule PIM1 inhibitors have been developed as potential therapeutic to reduce AD-neuropathology. This comprehensive study aims to address the activity of PIM1 inhibitor that has been tested for the treatment of AD, in addition to the pharmacological and structural aspects of PIM1.
Collapse
Affiliation(s)
- Shreyasi Meur
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, 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
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B.L Saha Road, Kolkata, 700053, West Bengal, India
| | - Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India.
| |
Collapse
|
2
|
Ren S, Sun C, Zhai W, Wei W, Liu J. Gaining new insights into the etiology of ulcerative colitis through a cross-tissue transcriptome-wide association study. Front Genet 2024; 15:1425370. [PMID: 39092429 PMCID: PMC11291327 DOI: 10.3389/fgene.2024.1425370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Background Genome-wide association studies (GWASs) have identified 38 loci associated with ulcerative colitis (UC) susceptibility, but the risk genes and their biological mechanisms remained to be comprehensively elucidated. Methods Multi-marker analysis of genomic annotation (MAGMA) software was used to annotate genes on GWAS summary statistics of UC from FinnGen database. Genetic analysis was performed to identify risk genes. Cross-tissue transcriptome-wide association study (TWAS) using the unified test for molecular signatures (UTMOST) was performed to compare GWAS summary statistics with gene expression matrix (from Genotype-Tissue Expression Project) for data integration. Subsequently, we used FUSION software to select key genes from the individual tissues. Additionally, conditional and joint analysis was conducted to improve our understanding on UC. Fine-mapping of causal gene sets (FOCUS) software was employed to accurately locate risk genes. The results of the four genetic analyses (MAGMA, UTMOST, FUSION and FOCUS) were combined to obtain a set of UC risk genes. Finally, Mendelian randomization (MR) analysis and Bayesian colocalization analysis were conducted to determine the causal relationship between the risk genes and UC. To test the robustness of our findings, the same approaches were taken to verify the GWAS data of UC on IEU. Results Multiple correction tests screened PIM3 as a risk gene for UC. The results of Bayesian colocalization analysis showed that the posterior probability of hypothesis 4 was 0.997 and 0.954 in the validation dataset. MR was conducted using the inverse variance weighting method and two single nucleotide polymorphisms (SNPs, rs28645887 and rs62231924) were included in the analysis (p < 0.001, 95%CI: 1.45-1.89). In the validation dataset, MR result was p < 0.001, 95%CI: 1.19-1.72, indicating a clear causal relationship between PIM3 and UC. Conclusion Our study validated PIM3 as a key risk gene for UC and its expression level may be related to the risk of UC, providing a novel reference for further improving the current understanding on the genetic structure of UC.
Collapse
Affiliation(s)
- Shijie Ren
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Chaodi Sun
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Wenjing Zhai
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Wenli Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jianping Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Department of Gastroenterology, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
Hochban PMM, Heyder L, Heine A, Diederich WE. What doesn't fit is made to fit: Pim-1 kinase adapts to the configuration of stilbene-based inhibitors. Arch Pharm (Weinheim) 2024; 357:e2400094. [PMID: 38631036 DOI: 10.1002/ardp.202400094] [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: 02/02/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024]
Abstract
Recently, we have developed novel Pim-1 kinase inhibitors starting from a dihydrobenzofuran core structure using a computational approach. Here, we report the design and synthesis of stilbene-based Pim-1 kinase inhibitors obtained by formal elimination of the dihydrofuran ring. These inhibitors of the first design cycle, which were obtained as inseparable cis/trans mixtures, showed affinities in the low single-digit micromolar range. To be able to further optimize these compounds in a structure-based fashion, we determined the X-ray structures of the protein-ligand-complexes. Surprisingly, only the cis-isomer binds upon crystallization of the cis/trans-mixture of the ligands with Pim-1 kinase and the substrate PIMTIDE, the binding mode being largely consistent with that predicted by docking. After crystallization of the exclusively trans-configured derivatives, a markedly different binding mode for the inhibitor and a concomitant rearrangement of the glycine-rich loop is observed, resulting in the ligand being deeply buried in the binding pocket.
Collapse
Affiliation(s)
- Phil M M Hochban
- Institut für Pharmazeutische Chemie, Zentrum für Tumor und Immunbiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Lukas Heyder
- Institut für Pharmazeutische Chemie, Zentrum für Tumor und Immunbiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Andreas Heine
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | - Wibke E Diederich
- Institut für Pharmazeutische Chemie, Zentrum für Tumor und Immunbiologie, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
4
|
Targeting Pim kinases in hematological cancers: molecular and clinical review. Mol Cancer 2023; 22:18. [PMID: 36694243 PMCID: PMC9875428 DOI: 10.1186/s12943-023-01721-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
Decades of research has recognized a solid role for Pim kinases in lymphoproliferative disorders. Often up-regulated following JAK/STAT and tyrosine kinase receptor signaling, Pim kinases regulate cell proliferation, survival, metabolism, cellular trafficking and signaling. Targeting Pim kinases represents an interesting approach since knock-down of Pim kinases leads to non-fatal phenotypes in vivo suggesting clinical inhibition of Pim may have less side effects. In addition, the ATP binding site offers unique characteristics that can be used for the development of small inhibitors targeting one or all Pim isoforms. This review takes a closer look at Pim kinase expression and involvement in hematopoietic cancers. Current and past clinical trials and in vitro characterization of Pim kinase inhibitors are examined and future directions are discussed. Current studies suggest that Pim kinase inhibition may be most valuable when accompanied by multi-drug targeting therapy.
Collapse
|
5
|
Reddy Peddi S, Kundenapally R, Kanth Sivan S, Somadi G, Manga V. A pragmatic pharmacophore informatics strategy to discover new potent inhibitors against pim-3. Struct Chem 2022. [DOI: 10.1007/s11224-022-01949-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Ismail MMF, Farrag AM, Abou‐El‐Ela D. Synthesis, anticancer screening, and in silico ADMEprediction of novel 2‐pyridonesas Pim inhibitors. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Magda M. F. Ismail
- Department of Pharmaceutical Chemistry, Faculty of PharmacyAl‐Azhar University Cairo Egypt
| | - Amel M. Farrag
- Department of Pharmaceutical Chemistry, Faculty of PharmacyAl‐Azhar University Cairo Egypt
| | - Dalal Abou‐El‐Ela
- Department of Pharmaceutical ChemistryFaculty of Pharmacy, Ain‐Shams University Cairo Egypt
| |
Collapse
|
7
|
Zhang S, Shuai L, Wang D, Huang T, Yang S, Miao M, Liu F, Xu J. Pim-1 Protects Retinal Ganglion Cells by Enhancing Their Regenerative Ability Following Optic Nerve Crush. Exp Neurobiol 2020; 29:249-272. [PMID: 32624507 PMCID: PMC7344373 DOI: 10.5607/en20019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 11/19/2022] Open
Abstract
Provirus integration site Moloney murine leukemia virus (Pim-1) is a proto-oncogene reported to be associated with cell proliferation, differentiation and survival. This study was to explore the neuroprotective role of Pim-1 in a rat model subjected to optic nerve crush (ONC), and discuss its related molecules in improving the intrinsic regeneration ability of retinal ganglion cells (RGCs). Immunofluorescence staining showed that AAV2- Pim-1 infected 71% RGCs and some amacrine cells in the retina. Real-time PCR and Western blotting showed that retina infection with AAV2- Pim-1 up-regulated the Pim-1 mRNA and protein expressions compared with AAV2-GFP group. Hematoxylin-Eosin (HE) staining, γ-synuclein immunohistochemistry, Cholera toxin B (CTB) tracing and TUNEL showed that RGCs transduction with AAV2-Pim-1 prior to ONC promoted the survival of damaged RGCs and decreased cell apoptosis. RITC anterograde labeling showed that Pim-1 overexpression increased axon regeneration and promoted the recovery of visual function by pupillary light reflex and flash visual evoked potential. Western blotting showed that Pim- 1 overexpression up-regulated the expression of Stat3, p-Stat3, Akt1, p-Akt1, Akt2 and p-Akt2, as well as βIII-tubulin, GAP-43 and 4E-BP1, and downregulated the expression of SOCS1 and SOCS3, Cleaved caspase 3, Bad and Bax. These results demonstrate that Pim-1 exerted a neuroprotective effect by promoting nerve regeneration and functional recovery of RGCs. In addition, it enhanced the intrinsic regeneration capacity of RGCs after ONC by activating Stat3, Akt1 and Akt2 pathways, and inhibiting the mitochondrial apoptosis pathways. These findings suggest that Pim-1 may prove to be a potential therapeutic target for the clinical treatment of optic nerve injury.
Collapse
Affiliation(s)
- Shoumei Zhang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Li Shuai
- Department of Health Administration, Second Military Medical University, Shanghai 200433, China
| | - Dong Wang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Tingting Huang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Shengsheng Yang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China
| | - Mingyong Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China
| | - Fang Liu
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Jiajun Xu
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
8
|
Ismail MM, Farrag AM, Harras MF, Ibrahim MH, Mehany AB. Apoptosis: A target for anticancer therapy with novel cyanopyridines. Bioorg Chem 2020; 94:103481. [DOI: 10.1016/j.bioorg.2019.103481] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
|
9
|
Wu W, Yu A, Chen K, Lu P, Yang J, Liu K, Mao Z, Yao Z. The Oncogene PIM1 Contributes to Cellular Senescence by Phosphorylating Staphylococcal Nuclease Domain-Containing Protein 1 (SND1). Med Sci Monit 2019; 25:8651-8659. [PMID: 31860636 PMCID: PMC6876065 DOI: 10.12659/msm.917867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background The oncogene PIM1, encoding a constitutively active serine/threonine protein kinase, is involved in the regulation of cell proliferation, survival, differentiation, and apoptosis. There is a growing body of literature on the role of PIM1-mediated cellular senescence, but the precise mechanism remains unclear. Material/Methods Silver staining and LC–MS/MS analysis were performed to investigate the protein interacting with PIM1. Immunofluorescence, Co-IP, and Western blot assay were used to assess the interaction of PIM1 and SND1. EdU incorporation and CCK8 assay were used to detect cell proliferation and immunohistochemistry was used to detect the level of the indicated protein. Results We found that PIM1 can bind directly and phosphorylate SND1. In addition, decreased expression of SND1 leads to the upregulation of SASP. SND1 is involved in cellular senescence induced by PIM1. Conclusions We investigated the role of PIM1 in oncogene-induced normal cellular senescence. Our results promote further understanding of the mechanisms underlying OIS and suggest potential applications for preventing tumorigenesis.
Collapse
Affiliation(s)
- Wu Wu
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China (mainland).,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (mainland)
| | - Aiqing Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (mainland)
| | - Keyu Chen
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China (mainland).,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (mainland)
| | - Peilin Lu
- Department of Dermatology, Sichuan Provincial Fourth People's Hospital, Chengdu, Sichuan, China (mainland)
| | - Jianming Yang
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China (mainland).,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (mainland)
| | - Kun Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (mainland)
| | - Zebin Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (mainland)
| | - Zhi Yao
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
10
|
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
|
11
|
Spirli A, Cheval L, Debonneville A, Penton D, Ronzaud C, Maillard M, Doucet A, Loffing J, Staub O. The serine-threonine kinase PIM3 is an aldosterone-regulated protein in the distal nephron. Physiol Rep 2019; 7:e14177. [PMID: 31397090 PMCID: PMC6687858 DOI: 10.14814/phy2.14177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/30/2022] Open
Abstract
The mineralocorticoid hormone aldosterone plays a crucial role in the control of Na+ and K+ balance, blood volume, and arterial blood pressure, by acting in the aldosterone-sensitive distal nephron (ASDN) and stimulating a complex transcriptional, translational, and cellular program. Because the complexity of the aldosterone response is still not fully appreciated, we aimed at identifying new elements in this pathway. Here, we demonstrate that the expression of the proto-oncogene PIM3 (Proviral Integration Site of Moloney Murine Leukemia Virus 3), a serine/threonine kinase belonging to the calcium/calmodulin-regulated group of kinases, is stimulated by aldosterone in vitro (mCCDcl1 cells), ex vivo (mouse kidney slices), and in vivo in mice. Characterizing a germline Pim3-/- mouse model, we found that these mice have an upregulated Renin-Angiotensin-Aldosterone System (RAAS), with high circulating aldosterone and plasma renin activity levels on both standard or Na+ -deficient diet. Surprisingly, we did not observe any obvious salt-losing phenotype in Pim3 KO mice as shown by normal blood pressure, plasma and urinary electrolytes, as well as unchanged expression levels of the major Na+ transport proteins. These observations suggest that the potential effects of the loss of the Pim3 gene are physiologically compensated. Indeed, the 2 other family members of the PIM kinase family, PIM1 and PIM2 are upregulated in the kidney of Pim3-/- mice, and may therefore be involved in such compensation. In conclusion, our data demonstrate that the PIM3 kinase is a novel aldosterone-induced protein, but its precise role in aldosterone-dependent renal homeostasis remains to be determined.
Collapse
Affiliation(s)
- Alessia Spirli
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| | - Lydie Cheval
- Centre de Recherche des CordeliersINSERM, Sorbonne Universités, USPC, Université Paris Descartes, Université Paris Diderot, Physiologie Rénale et TubulopathiesParisFrance
| | - Anne Debonneville
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| | - David Penton
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
- Institute of AnatomyUniversity of ZurichZurichSwitzerland
| | - Caroline Ronzaud
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| | - Marc Maillard
- Service of NephrologyLausanne University Hospital (CHUV)LausanneSwitzerland
| | - Alain Doucet
- Centre de Recherche des CordeliersINSERM, Sorbonne Universités, USPC, Université Paris Descartes, Université Paris Diderot, Physiologie Rénale et TubulopathiesParisFrance
| | - Johannes Loffing
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
- Institute of AnatomyUniversity of ZurichZurichSwitzerland
| | - Olivier Staub
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| |
Collapse
|
12
|
Wang G, Liu G, Ye Y, Fu Y, Zhang X. Bufothionine exerts anti-cancer activities in gastric cancer through Pim3. Life Sci 2019; 232:116615. [PMID: 31260686 DOI: 10.1016/j.lfs.2019.116615] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/19/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022]
Abstract
AIM Gastric cancer (GC) is the fourth most common cancer globally. Bufothionine is a major active constituent of Cinobufacini (Huachansu), which is extracted from the skin and parotid venom gland of the toad Bufo bufo gargarizans Cantor. It exhibits anti-cancer activities in vitro. However, whether bufothionine exerts anti-cancer activities against GC is unknown. This study was designed to evaluate the efficacy of bufothionine in vitro and in vivo. MATERIAL AND METHODS MKN28 and AGS cells were chosen as cell models to study the anti-cancer effect of bufothionine. Cell viability was determined by CCK-8 assay, while the effect of bufothionine on cell membrane integrity was examined by LDH assay. Cell apoptosis was detected by Hoechst/PI staining and Annexin V-FITC/PI staining followed by flow cytometry analysis. The expression levels of proteins involved were examined using western blotting. I-Traq analysis was conducted to identify the differentially expressed genes in AGS cells following bufothionine treatment. The anti-growth effect of bufothionine was validated in vivo using a GC xenograft model. KEY FINDINGS The results revealed that bufothionine prevented the growth, destroyed cell membrane and promoted apoptotic cell death of GC cells. iTRAQ analysis revealed thatPIM3 might be a molecular target responsible for the anti-cancer effects of bufothionine. It was also found that PIM3 knockdown significantly augmented the anti-growth and pro-apoptotic effects of bufothionine in GC cells. In contrast, ectopic PIM3 expression markedly dampened the anti-neoplastic activities of bufothionine. The expression of PIM3 was also suppressed by bufothionine treatment in xenograft tumor tissue. SIGNIFICANCE Bufothionine exhibited anti-cancer activities in vitro and in vivo in GC via downregulating PIM3.
Collapse
Affiliation(s)
- Guojun Wang
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Guanghui Liu
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yanwei Ye
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yang Fu
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiefu Zhang
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
13
|
Zhao L, Yuan X, Wang J, Feng Y, Ji F, Li Z, Bian J. A review on flavones targeting serine/threonine protein kinases for potential anticancer drugs. Bioorg Med Chem 2019; 27:677-685. [DOI: 10.1016/j.bmc.2019.01.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
|
14
|
Qi Q, Pan Y, Han S, Liao H, Jiang Y, Shen J, Zhong L, Wang X, Chen J. PIM3 Functions as Oncogenic Factor and Promotes the Tumor Growth and Metastasis in Colorectal Cancer. Anat Rec (Hoboken) 2018; 302:1552-1560. [PMID: 30417983 DOI: 10.1002/ar.24024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/28/2018] [Accepted: 08/23/2018] [Indexed: 12/26/2022]
Abstract
Colorectal cancer (CRC) is one of the common human malignancies. Discovery and identification of novel therapeutic target is imperative to improve the prognosis of CRC patients. As a member of the PIM family, PIM3 has been found to be overexpressed in a variety of cancerous tumors. In this study, we evaluated the expression of PIM3 in CRC tissues and analyzed the role of PIM3 in CRC. Our results showed that PIM3 expression was significantly higher in CRC tissues compared with adjacent noncancerous tissues. The PIM3 expression level was found to be correlated with advanced disease stage and lymph node metastasis. Moreover, PIM3 was found to be able to predict poor prognosis in CRC patients as an independent factor. In vitro studies also showed that knockdown of PIM3 exhibited inhibitory effect on cell growth, promoted cell apoptosis and dampened invasive capability of HCT116 and SW620 cells. Moreover, PIM3 knockdown was able to delay tumor growth and suppress lung metastasis in xenograft model. Our results indicated that PIM3 is a potential therapeutic target for CRC. Anat Rec, 302:1552-1560, 2019. © 2018 American Association for Anatomy.
Collapse
Affiliation(s)
- Quan Qi
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yuefen Pan
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang, China
| | - Shuwen Han
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang, China
| | - Haihong Liao
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang, China
| | - Yizhen Jiang
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang, China
| | - Junjun Shen
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang, China
| | - Liping Zhong
- Department of Medical Oncology, Huzhou Central Hospital, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Jie Chen
- Department of Gastroenterology, Huzhou Central Hospital, Zhejiang, China
| |
Collapse
|
15
|
Zhang S, Wang D, Huang T, Liu F, Shuai L, Xu J. Pim-1 Expression in Rat Retina and its Changes after Optic Nerve Crush. Anat Rec (Hoboken) 2018; 301:1968-1976. [PMID: 30299595 DOI: 10.1002/ar.23947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 01/19/2018] [Accepted: 02/16/2018] [Indexed: 11/11/2022]
Abstract
Pim-1 is a proto-oncogene which has been discovered to involve in cell proliferation, differentiation, and survival. In this study, we observed the expression of Pim-1 in neonatal and adult rat retina and the changes in rat retina following optic nerve crush (ONC) in order to explore the relationship between Pim-1 and the survival of retinal ganglion cells (RGC). We discovered that Pim-1 was distributed mainly in retinal pigment epithelial cells (RPE) and retinal ganglion cell layer (GCL) in normal newborn rats, and it appeared in RPE, cone rod cell layer and GCL in normal adult rats by immunohistochemistry. Our double immunofluorescent staining of Pim-1 and γ-synuclein further confirmed that Pim-1 was localized in 80% of RGC. Moreover, we found that the amount of Pim-1 mRNA and protein in adult rat retina was transiently increased after ONC and then decreased 2 weeks after ONC, and the expression level was lower than that of neonatal rat retina under all conditions. We also discovered that Pim-1 expression in GCL detected by immunohistochemistry was upregulated at Day 1 and Day 3 after ONC, but downregulated at Day 14 after ONC when the survival of RGC was decreased and the apoptotic cells in GCL were increased by hematoxylin-eosin staining, immunohistochemistry, and TUNEL detection. We suggest that the overexpression of Pim-1 in the RGC is related to the optic nerve repair while the low expression of Pim-1 in RGC may be associated with apoptosis and weak intrinsic regeneration ability of RGC. Anat Rec, 301:1968-1976, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shoumei Zhang
- Department of Anatomy, Second Military Medical University, Shanghai, China
| | - Dong Wang
- Department of Anatomy, Second Military Medical University, Shanghai, China
| | - Tingting Huang
- Department of Anatomy, Second Military Medical University, Shanghai, China
| | - Fang Liu
- Department of Anatomy, Second Military Medical University, Shanghai, China
| | - Li Shuai
- Department of Health Administration, Second Military Medical University, Shanghai, China
| | - Jiajun Xu
- Department of Anatomy, Second Military Medical University, Shanghai, China
| |
Collapse
|
16
|
Juszczak GR, Stankiewicz AM. Glucocorticoids, genes and brain function. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:136-168. [PMID: 29180230 DOI: 10.1016/j.pnpbp.2017.11.020] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/18/2017] [Accepted: 11/23/2017] [Indexed: 01/02/2023]
Abstract
The identification of key genes in transcriptomic data constitutes a huge challenge. Our review of microarray reports revealed 88 genes whose transcription is consistently regulated by glucocorticoids (GCs), such as cortisol, corticosterone and dexamethasone, in the brain. Replicable transcriptomic data were combined with biochemical and physiological data to create an integrated view of the effects induced by GCs. The most frequently reported genes were Errfi1 and Ddit4. Their up-regulation was associated with the altered transcription of genes regulating growth factor and mTORC1 signaling (Gab1, Tsc22d3, Dusp1, Ndrg2, Ppp5c and Sesn1) and progression of the cell cycle (Ccnd1, Cdkn1a and Cables1). The GC-induced reprogramming of cell function involves changes in the mRNA level of genes responsible for the regulation of transcription (Klf9, Bcl6, Klf15, Tle3, Cxxc5, Litaf, Tle4, Jun, Sox4, Sox2, Sox9, Irf1, Sall2, Nfkbia and Id1) and the selective degradation of mRNA (Tob2). Other genes are involved in the regulation of metabolism (Gpd1, Aldoc and Pdk4), actin cytoskeleton (Myh2, Nedd9, Mical2, Rhou, Arl4d, Osbpl3, Arhgef3, Sdc4, Rdx, Wipf3, Chst1 and Hepacam), autophagy (Eva1a and Plekhf1), vesicular transport (Rhob, Ehd3, Vps37b and Scamp2), gap junctions (Gjb6), immune response (Tiparp, Mertk, Lyve1 and Il6r), signaling mediated by thyroid hormones (Thra and Sult1a1), calcium (Calm2), adrenaline/noradrenaline (Adcy9 and Adra1d), neuropeptide Y (Npy1r) and histamine (Hdc). GCs also affected genes involved in the synthesis of polyamines (Azin1) and taurine (Cdo1). The actions of GCs are restrained by feedback mechanisms depending on the transcription of Sgk1, Fkbp5 and Nr3c1. A side effect induced by GCs is increased production of reactive oxygen species. Available data show that the brain's response to GCs is part of an emergency mode characterized by inactivation of non-core activities, restrained inflammation, restriction of investments (growth), improved efficiency of energy production and the removal of unnecessary or malfunctioning cellular components to conserve energy and maintain nutrient supply during the stress response.
Collapse
Affiliation(s)
- Grzegorz R Juszczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland.
| | - Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland
| |
Collapse
|
17
|
Identification of 22q13 genes most likely to contribute to Phelan McDermid syndrome. Eur J Hum Genet 2018; 26:293-302. [PMID: 29358616 PMCID: PMC5838980 DOI: 10.1038/s41431-017-0042-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/04/2017] [Accepted: 10/31/2017] [Indexed: 01/02/2023] Open
Abstract
Chromosome 22q13.3 deletion (Phelan McDermid) syndrome (PMS) is a rare genetic neurodevelopmental disorder resulting from deletions or other genetic variants on distal 22q. Pathological variants of the SHANK3 gene have been identified, but terminal chromosomal deletions including SHANK3 are most common. Terminal deletions disrupt up to 108 protein-coding genes. The impact of these losses is highly variable and includes both significantly impairing neurodevelopmental and somatic manifestations. The current review combines two metrics, prevalence of gene loss and predicted loss pathogenicity, to identify likely contributors to phenotypic expression. These genes are grouped according to function as follows: molecular signaling at glutamate synapses, phenotypes involving neuropsychiatric disorders, involvement in multicellular organization, cerebellar development and functioning, and mitochondrial. The likely most impactful genes are reviewed to provide information for future clinical and translational investigations.
Collapse
|
18
|
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]
|
19
|
Amin AD, Peters TL, Li L, Rajan SS, Choudhari R, Puvvada SD, Schatz JH. Diffuse large B-cell lymphoma: can genomics improve treatment options for a curable cancer? Cold Spring Harb Mol Case Stud 2017; 3:a001719. [PMID: 28487884 PMCID: PMC5411687 DOI: 10.1101/mcs.a001719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gene-expression profiling and next-generation sequencing have defined diffuse large B-cell lymphoma (DLBCL), the most common lymphoma diagnosis, as a heterogeneous group of subentities. Despite ongoing explosions of data illuminating disparate pathogenic mechanisms, however, the five-drug chemoimmunotherapy combination R-CHOP remains the frontline standard treatment. This has not changed in 15 years, since the anti-CD20 monoclonal antibody rituximab was added to the CHOP backbone, which first entered use in the 1970s. At least a third of patients are not cured by R-CHOP, and relapsed or refractory DLBCL is fatal in ∼90%. Targeted small-molecule inhibitors against distinct molecular pathways activated in different subgroups of DLBCL have so far translated poorly into the clinic, justifying the ongoing reliance on R-CHOP and other long-established chemotherapy-driven combinations. New drugs and improved identification of biomarkers in real time, however, show potential to change the situation eventually, despite some recent setbacks. Here, we review established and putative molecular drivers of DLBCL identified through large-scale genomics, highlighting among other things the care that must be taken when differentiating drivers from passengers, which is influenced by the promiscuity of activation-induced cytidine deaminase. Furthermore, we discuss why, despite having so much genomic data available, it has been difficult to move toward personalized medicine for this umbrella disorder and some steps that may be taken to hasten the process.
Collapse
Affiliation(s)
- Amit Dipak Amin
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Tara L Peters
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Lingxiao Li
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Soumya Sundara Rajan
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ramesh Choudhari
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Soham D Puvvada
- Department of Medicine, Division of Hematology-Oncology, University of Arizona Comprehensive Cancer Center, Tucson, Arizona 85719, USA
| | - Jonathan H Schatz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
20
|
Chang W, Liu M, Xu J, Fu H, Zhou B, Yuan T, Chen P. MiR-377 inhibits the proliferation of pancreatic cancer by targeting Pim-3. Tumour Biol 2016; 37:14813-14824. [PMID: 27638830 DOI: 10.1007/s13277-016-5295-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/31/2016] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) play important roles in the regulation of various tumor biological processes including proliferation and apoptosis. MiR-377 has been implicated in many types of cancer, whereas its expressional feature and potential biological function in pancreatic ductal adenocarcinoma (PDAC) remains unclear. In this study, we scanned the global miRNA expression profiles in PDAC from The Cancer Genome Atlas (TCGA) and found miR-377 was down-regulated significantly in PDAC. Then, its expression was measured in both pancreatic cancer tissues and cells; the data showed that miR-377 was de-regulated and inversely correlated with pathologic parameters of tumor growth or metastasis. We generated PDAC cell lines with stable overexpression or inhibition of miR-377, and our results indicated that miR-377 up-regulation significantly promoted cell viability, proliferation, and migration in PDAC cells, and also induced cell apoptosis and cell cycle arrest simultaneously. Binding-site predictions by bioinformatics showed that Pim-3 might be a potential target of miR-377. Luciferase reporter assay ulteriorly identified that miR-377 suppressed Pim-3 expression by binding the 3'-UTR. In tumor tissues, we also showed that the Pim-3 expression was inversely correlated with that of miR-377. Furthermore, stable ectopic miR-377 expression in pancreatic cancer cell lines suppressed Pim-3 expression, leading to the attenuation of Bad phosphorylation level at its Ser112 and promoting cell apoptosis. Overall, these results reveal that miR-377 may have tumor growth suppression function by down-regulating Pim-3 kinase expression to inhibit both pancreatic tumor growth and migration, and induce cell apoptosis. Hence, miR-377 may be a potential diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Weihua Chang
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China
| | - Menggang Liu
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China
| | - Jianhua Xu
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China
| | - Hangwei Fu
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China
| | - Bo Zhou
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China
| | - Tao Yuan
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China.
| | - Ping Chen
- Department of Hepatobiliary Surgery, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, No.10, Changjiangzhilu Road, Yu Zhong District, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
21
|
Zhuang H, Zhao MY, Hei KW, Yang BC, Sun L, Du X, Li YM. Aberrant expression of pim-3 promotes proliferation and migration of ovarian cancer cells. Asian Pac J Cancer Prev 2016; 16:3325-31. [PMID: 25921139 DOI: 10.7314/apjcp.2015.16.8.3325] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Pim kinase-3(Pim-3), a member of serine/threonine protein kinases, has been implicated in multiple human cancers and involved in Myc-induced tumorigenesis. However, little is known regarding its expression and biological function in human ovarian cancer. In this study we showed that the clinical significance and biological functions of Pim-3 in ovarian cancer and found that higher Pim-3 mRNA level are detected in ovarian cancer tissues than those in normal ovarian tissues. There are significant correlations between higher Pim-3 expression levels with the FIGO stage, histopathological subtypes, and distant metastasis in ovarian cancer patients. Lentivirus-mediated gene overexpression of Pim-3 significantly promotes the proliferation and migration of SKOV3 cell lines. Furthermore, MACC1 and Pim-3 expression were significantly correlated in human ovarian cancer cells, and overexpression of Pim-3 in ovary cancer cells increased MACC1 mRNA and protein expression. The data indicate that Pim-3 acts as a putative oncogene in ovary cancer and could be a viable diagnostic and therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Hao Zhuang
- Department of Medical Microbiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China E-mail : ,
| | | | | | | | | | | | | |
Collapse
|
22
|
DU JUNDONG, ZHENG XI, CAI SHOUWANG, ZHU ZIMAN, TAN JINGWANG, HU BIN, HUANG ZHIQIANG, JIAO HUABO. MicroRNA-506 participates in pancreatic cancer pathogenesis by targeting PIM3. Mol Med Rep 2015; 12:5121-6. [DOI: 10.3892/mmr.2015.4109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 03/18/2015] [Indexed: 11/06/2022] Open
|
23
|
Mondello P, Cuzzocrea S, Mian M. Pim kinases in hematological malignancies: where are we now and where are we going? J Hematol Oncol 2014; 7:95. [PMID: 25491234 PMCID: PMC4266197 DOI: 10.1186/s13045-014-0095-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022] Open
Abstract
The proviral insertion in murine (PIM) lymphoma proteins are a serine/threonine kinase family composed of three isoformes: Pim-1, Pim-2 and Pim-3. They play a critical role in the control of cell proliferation, survival, homing and migration. Recently, overexpression of Pim kinases has been reported in human tumors, mainly in hematologic malignancies. In vitro and in vivo studies have confirmed their oncogenic potential. Indeed, PIM kinases have shown to be involved in tumorgenesis, to enhance tumor growth and to induce chemo-resistance, which is why they have become an attractive therapeutic target for cancer therapy. Novel molecules inhibiting Pim kinases have been evaluated in preclinical studies, demonstrating to be effective and with a favorable toxicity profile. Given the promising results, some of these compounds are currently under investigation in clinical trials. Herein, we provide an overview of the biological activity of PIM-kinases, their role in hematologic malignancies and future therapeutic opportunities.
Collapse
Affiliation(s)
- Patrizia Mondello
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125, Messina, Italy. .,Department of Biological and Environmental Sciences, University of Messina, Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy.
| | - Michael Mian
- Department of Hematology, Hospital S. Maurizio, Bolzano/Bozen, Italy. .,Department of Internal Medicine V, Hematology & Oncology, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
24
|
Foulks JM, Carpenter KJ, Luo B, Xu Y, Senina A, Nix R, Chan A, Clifford A, Wilkes M, Vollmer D, Brenning B, Merx S, Lai S, McCullar MV, Ho KK, Albertson DJ, Call LT, Bearss JJ, Tripp S, Liu T, Stephens BJ, Mollard A, Warner SL, Bearss DJ, Kanner SB. A small-molecule inhibitor of PIM kinases as a potential treatment for urothelial carcinomas. Neoplasia 2014; 16:403-12. [PMID: 24953177 PMCID: PMC4198696 DOI: 10.1016/j.neo.2014.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 11/30/2022]
Abstract
The proto-oncogene proviral integration site for moloney murine leukemia virus (PIM) kinases (PIM-1, PIM-2, and PIM-3) are serine/threonine kinases that are involved in a number of signaling pathways important to cancer cells. PIM kinases act in downstream effector functions as inhibitors of apoptosis and as positive regulators of G1-S phase progression through the cell cycle. PIM kinases are upregulated in multiple cancer indications, including lymphoma, leukemia, multiple myeloma, and prostate, gastric, and head and neck cancers. Overexpression of one or more PIM family members in patient tumors frequently correlates with poor prognosis. The aim of this investigation was to evaluate PIM expression in low- and high-grade urothelial carcinoma and to assess the role PIM function in disease progression and their potential to serve as molecular targets for therapy. One hundred thirty-seven cases of urothelial carcinoma were included in this study of surgical biopsy and resection specimens. High levels of expression of all three PIM family members were observed in both noninvasive and invasive urothelial carcinomas. The second-generation PIM inhibitor, TP-3654, displays submicromolar activity in pharmacodynamic biomarker modulation, cell proliferation studies, and colony formation assays using the UM-UC-3 bladder cancer cell line. TP-3654 displays favorable human ether-à-go-go-related gene and cytochrome P450 inhibition profiles compared with the first-generation PIM inhibitor, SGI-1776, and exhibits oral bioavailability. In vivo xenograft studies using a bladder cancer cell line show that PIM kinase inhibition can reduce tumor growth, suggesting that PIM kinase inhibitors may be active in human urothelial carcinomas.
Collapse
Affiliation(s)
| | | | - Bai Luo
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | - Yong Xu
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | - Anna Senina
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | - Rebecca Nix
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | - Ashley Chan
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | | | | | | | | | | | - Shuping Lai
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | | | - Koc-Kan Ho
- Astex Pharmaceuticals, Inc, Salt Lake City, UT
| | - Daniel J Albertson
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | | | - Jared J Bearss
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - Ting Liu
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | | | | | | | | | | |
Collapse
|
25
|
Liu B, Wang Z, Li HY, Zhang B, Ping B, Li YY. Pim-3 promotes human pancreatic cancer growth by regulating tumor vasculogenesis. Oncol Rep 2014; 31:2625-34. [PMID: 24789328 DOI: 10.3892/or.2014.3158] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/08/2014] [Indexed: 11/05/2022] Open
Abstract
Pim-3, a proto-oncogene with serine/threonine kinase activity, is aberrantly expressed in malignant lesions, but not in normal pancreatic tissues. To assess the role of Pim-3 in human pancreatic carcinogenesis in vivo and to determine the underlying Pim-3 signaling regulatory mechanisms, we established MiaPaca-2 cells overexpressing wild-type Pim-3 or Pim-3 kinase dead mutants (K69M-Pim-3) as well as PCI55 cells stably expressing Pim-3 shRNA or scrambled shRNA in a tetracycline-inducible manner. In addition, we conducted studies utilizing a nude mouse tumor xenograft model. Our results demonstrated that cells stably overexpressing wild-type Pim-3 exhibited functionally enhanced phosphorylation of Bad at Ser112 and increased proliferation. In contrast, the stable inactivation of Pim-3 by K69M-Pim-3 or silencing of Pim-3 expression by Pim-3 shRNA resulted in functionally decreased phosphorylation of Bad at Ser112 and higher apoptotic cells. Following subcutaneous injection of these stable cell lines, nude mice injected with Pim-3 overexpressing cells developed 100% subcutaneous tumors, together with increased PCNA-positive cells and enhanced intratumoral CD31-positive vascular areas. On the other hand, intratumoral neovascularization and tumor cell proliferation was attenuated in mice injected with Pim-3 kinase inactive cells, eventually reducing tumorigenicity in these mice to 46.6%. Moreover, Pim-3 overexpression upregulated the intratumoral levels of pSTAT3Try705, pSurvivinThr34, HGF, EGF, FGF-2 and VEGF, while the increases were markedly diminished on Pim-3 kinase inactivation. Collectively, the Pim-3 kinase emerges as being involved in accelerating human pancreatic cancer development and in promoting tumor neovascularization and subsequent tumor growth. Targeting Pim-3 may play a dual role in halting tumor progression, by promoting tumor cell death and blocking angiogenesis.
Collapse
Affiliation(s)
- Bin Liu
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Zhen Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Hong-Yu Li
- Department of Gastroenterology, Shenyang General Hospital, Shenyang, Liaoning, P.R. China
| | - Bo Zhang
- Department of Pancreas and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Bo Ping
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ying-Yi Li
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
26
|
Xu D, Cobb MG, Gavilano L, Witherspoon SM, Williams D, White CD, Taverna P, Bednarski BK, Kim HJ, Baldwin AS, Baines AT. Inhibition of oncogenic Pim-3 kinase modulates transformed growth and chemosensitizes pancreatic cancer cells to gemcitabine. Cancer Biol Ther 2014; 14:492-501. [PMID: 23760491 DOI: 10.4161/cbt.24343] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a 5-year survival rate of only 6%. Although the cytosine analog gemcitabine is the drug commonly used to treat PDAC, chemoresistance unfortunately renders the drug ineffective. Thus, strategies that can decrease this resistance will be essential for improving the dismal outcome of patients suffering from this disease. We previously observed that oncogenic Pim-1 kinase was aberrantly expressed in PDAC tissues and cell lines and was responsible for radioresistance. Furthermore, members of the Pim family have been shown to reduce the efficacy of chemotherapeutic drugs in cancer. Therefore, we attempted to evaluate the role of Pim-3 in chemoresistance of PDAC cells. We were able to confirm upregulation of the Pim-3 oncogene in PDAC tissues and cell lines versus normal samples. Biological consequences of inhibiting Pim-3 expression with shRNA-mediated suppression included decreases in anchorage-dependent growth, invasion through Matrigel and chemoresistance to gemcitabine as measured by caspase-3 activity. Additionally, we were able to demonstrate that Pim-1 and Pim-3 play overlapping but non-identical roles as it relates to gemcitabine sensitivity of pancreatic cancer cells. To further support the role of Pim-3 suppression in sensitizing PDAC cells to gemcitabine, we used the pharmacological Pim kinase inhibitor SGI-1776. Treatment of PDAC cells with SGI-1776 resulted in decreased phosphorylation of the proapoptotic protein Bad and cell cycle changes. When SGI-1776 was combined with gemcitabine, there was a greater decrease in cell viability in the PDAC cells versus cells treated with either of the drugs separately. These results suggest combining drug therapies that inhibit Pim kinases, such as Pim-3, with chemotherapeutic agents, to aid in decreasing chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Dapeng Xu
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang C, Li HY, Liu B, Huang S, Wu L, Li YY. Pim-3 promotes the growth of human pancreatic cancer in the orthotopic nude mouse model through vascular endothelium growth factor. J Surg Res 2013; 185:595-604. [PMID: 23845873 DOI: 10.1016/j.jss.2013.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/28/2013] [Accepted: 06/05/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND As one of the most lethal cancers, pancreatic cancer presents poor prognosis with an overall 5-y survival of less than 5%. We previously reported that Pim-3, a member of the proto-oncogene Pim family that encodes serine/threonine kinases, is aberrantly expressed in human pancreatic cancer lesions. In the current study, we investigated the role of Pim-3 in promoting tumor growth and angiogenesis in an orthotopic nude mouse model of human pancreatic cancer. METHODS We constructed retroviral vectors for human Pim-3 and a kinase-dead mutant of human Pim-3 (K69M); the retroviral supernatants generated from these vectors were then used to infect the human pancreatic cancer cell line MiaPaCa-2 to establish stable cell lines. We assessed cell proliferation using CCK-8, tumor growth, and angiogenesis in vivo in an orthotopic mouse model of pancreatic cancer. While tumor size was measured using magnetic resonance imaging, the tumor tissues were excised for protein extraction and histological analysis to detect vascular endothelium growth factor (VEGF) expression and vessel density. RESULTS We established an orthotopic nude mouse model of human pancreatic cancer. We observed that Pim-3 promoted the proliferation of human pancreatic cancer cells, both in vitro and in vivo. Moreover, Pim-3 is required for vasculogenesis of primary human pancreatic tumors in vivo and promotion of angiogenesis through the induction of VEGF expression. CONCLUSIONS Pim-3 can promote tumor growth and angiogenesis by stimulating the VEGF pathway.
Collapse
Affiliation(s)
- Chen Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
28
|
Tahvanainen J, Kyläniemi MK, Kanduri K, Gupta B, Lähteenmäki H, Kallonen T, Rajavuori A, Rasool O, Koskinen PJ, Rao KVS, Lähdesmäki H, Lahesmaa R. Proviral integration site for Moloney murine leukemia virus (PIM) kinases promote human T helper 1 cell differentiation. J Biol Chem 2012; 288:3048-58. [PMID: 23209281 PMCID: PMC3561529 DOI: 10.1074/jbc.m112.361709] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The differentiation of human primary T helper 1 (Th1) cells from naïve precursor cells is regulated by a complex, interrelated signaling network. The identification of factors regulating the early steps of Th1 cell polarization can provide important insight in the development of therapeutics for many inflammatory and autoimmune diseases. The serine/threonine-specific proviral integration site for Moloney murine leukemia virus (PIM) kinases PIM1 and PIM2 have been implicated in the cytokine-dependent proliferation and survival of lymphocytes. We have established that the third member of this family, PIM3, is also expressed in human primary Th cells and identified a new function for the entire PIM kinase family in T lymphocytes. Although PIM kinases are expressed more in Th1 than Th2 cells, we demonstrate here that these kinases positively influence Th1 cell differentiation. Our RNA interference results from human primary Th cells also suggest that PIM kinases promote the production of IFNγ, the hallmark cytokine produced by Th1 cells. Consistent with this, they also seem to be important for the up-regulation of the critical Th1-driving factor, T box expressed in T cells (T-BET), and the IL-12/STAT4 signaling pathway during the early Th1 differentiation process. In summary, we have identified PIM kinases as new regulators of human primary Th1 cell differentiation, thus providing new insights into the mechanisms controlling the selective development of human Th cell subsets.
Collapse
Affiliation(s)
- Johanna Tahvanainen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, 20520 Turku, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Walpen T, Kalus I, Schwaller J, Peier MA, Battegay EJ, Humar R. Nuclear PIM1 confers resistance to rapamycin-impaired endothelial proliferation. Biochem Biophys Res Commun 2012; 429:24-30. [PMID: 23131564 DOI: 10.1016/j.bbrc.2012.10.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 10/24/2012] [Indexed: 01/15/2023]
Abstract
The PIM serine/threonine kinases and the mTOR/AKT pathway integrate growth factor signaling and promote cell proliferation and survival. They both share phosphorylation targets and have overlapping functions, which can partially substitute for each other. In cancer cells PIM kinases have been reported to produce resistance to mTOR inhibition by rapamycin. Tumor growth depends highly on blood vessel infiltration into the malignant tissue and therefore on endothelial cell proliferation. We therefore investigated how the PIM1 kinase modulates growth inhibitory effects of rapamycin in mouse aortic endothelial cells (MAEC). We found that proliferation of MAEC lacking Pim1 was significantly more sensitive to rapamycin inhibition, compared to wildtype cells. Inhibition of mTOR and AKT in normal MAEC resulted in significantly elevated PIM1 protein levels in the cytosol and in the nucleus. We observed that truncation of the C-terminal part of Pim1 beyond Ser 276 resulted in almost exclusive nuclear localization of the protein. Re-expression of this Pim1 deletion mutant significantly increased the proliferation of Pim1(-/-) cells when compared to expression of the wildtype Pim1 cDNA. Finally, overexpression of the nuclear localization mutant and the wildtype Pim1 resulted in complete resistance to growth inhibition by rapamycin. Thus, mTOR inhibition-induced nuclear accumulation of PIM1 or expression of a nuclear C-terminal PIM1 truncation mutant is sufficient to increase endothelial cell proliferation, suggesting that nuclear localization of PIM1 is important for resistance of MAEC to rapamycin-mediated inhibition of proliferation.
Collapse
Affiliation(s)
- Thomas Walpen
- Research Unit, Division Internal Medicine, University Hospital Zürich, 8091 Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
30
|
Ogawa N, Yuki H, Tanaka A. Insights from Pim1 structure for anti-cancer drug design. Expert Opin Drug Discov 2012; 7:1177-92. [DOI: 10.1517/17460441.2012.727394] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
31
|
Abstract
The PIM genes represent a family of proto-oncogenes that encode three different serine/threonine protein kinases (PIM1, PIM2 and PIM3) with essential roles in the regulation of signal transduction cascades, which promote cell survival, proliferation and drug resistance. PIM kinases are overexpressed in several hematopoietic tumors and support in vitro and in vivo malignant cell growth and survival, through cell cycle regulation and inhibition of apoptosis. PIM kinases do not have an identified regulatory domain, which means that these proteins are constitutively active once transcribed. They appear to be critical downstream effectors of important oncoproteins and, when overexpressed, can mediate drug resistance to available agents, such as rapamycin. Recent crystallography studies reveal that, unlike other kinases, they possess a hinge region, which creates a unique binding pocket for ATP, offering a target for an increasing number of potent small-molecule PIM kinase inhibitors. Preclinical studies in models of various hematologic cancers indicate that these novel agents show promising activity and some of them are currently being evaluated in a clinical setting. In this review, we profile the PIM kinases as targets for therapeutics in hematologic malignancies.
Collapse
Affiliation(s)
- Yesid Alvarado
- Department of Hematology/Oncology, Cancer Therapy & Research Center, The University of Texas Health Science Center San Antonio, 7979 Wurzbach Road, MC8232, San Antonio, 78229, TX, USA
| | | | | |
Collapse
|
32
|
Walpen T, Peier M, Haas E, Kalus I, Schwaller J, Battegay E, Humar R. Loss ofPim1Imposes a Hyperadhesive Phenotype on Endothelial Cells. Cell Physiol Biochem 2012. [DOI: 10.1159/000341484] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
33
|
Zhukova YN, Alekseeva MG, Zakharevich NV, Shtil AA, Danilenko VN. Pim family of protein kinases: Structure, functions, and roles in hematopoietic malignancies. Mol Biol 2011. [DOI: 10.1134/s0026893311040170] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
Abstract
Pim-3 is a member of the Provirus integrating site Moloney murine leukemia virus (Pim) family, which belongs to the Ca(2+) /calmodulin-dependent protein kinase (CaMK) group and exhibits serine/threonine kinase activity. Similar to other members of the Pim family (i.e. Pim-1 and Pim-2), Pim-3 can prevent apoptosis and promote cell survival and protein translation, thereby enhancing cell proliferation of normal and malignant cells. Pim-3 is expressed in vital organs, such as the heart, lung, and brain. However, minimal phenotypic changes in Pim-3-deficient mice suggest that Pim-3 may be physiologically dispensable. Pim-3 expression is enhanced in several cancer tissues, particularly those of endoderm-derived organs, including the liver, pancreas, colon, and stomach. The development of hepatocellular carcinoma is accelerated in mice expressing the Pim-3 gene selectively in the liver only when these mice are treated with a hepatocarcinogen, indicating that Pim-3 can act as a promoter but not as an initiator. Moreover, inhibition of Pim-3 expression can retard in vitro cell proliferation of hepatocellular, pancreatic, and colon carcinoma cell lines by promoting cell apoptosis. Furthermore, a Pim-3 kinase inhibitor has been reported to inhibit cell proliferation in an in vivo xenograft model using a human pancreatic cancer cell line without inducing any major adverse effects. Thus, Pim-3 kinase may be a candidate molecule for the development of molecular targeting drugs against cancer.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Microenvironment Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
| | | | | |
Collapse
|
35
|
Isaac M, Siu A, Jongstra J. The oncogenic PIM kinase family regulates drug resistance through multiple mechanisms. Drug Resist Updat 2011; 14:203-11. [DOI: 10.1016/j.drup.2011.04.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 04/18/2011] [Accepted: 04/18/2011] [Indexed: 01/05/2023]
|
36
|
Abstract
Pim oncogenes are overexpressed in a wide range of tumours from a haematological and epithelial origin. Pim genes encode serine/threonine kinases that have been shown to counteract the increased sensitivity to apoptosis induction that is associated with MYC-driven tumorigenesis. Recently, considerable progress has been made in characterizing the pathways of PIM-mediated survival signalling. Given the unique structure of their active site and the minimal phenotype of mice mutant for all Pim family members, these oncogenes might be promising targets for highly specific and selective drugs with favourable toxicity profiles. In this Review, we discuss the physiological functions and oncogenic activities of Pim kinases.
Collapse
Affiliation(s)
- Martijn C Nawijn
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | | |
Collapse
|
37
|
Zhao L, Wang Y, Min X, Yang H, Zhang P, Zeng Q. Ischemia-reperfusion injury up-regulates Pim-3 gene expression in myocardial tissue. ACTA ACUST UNITED AC 2010; 30:704-8. [PMID: 21181358 DOI: 10.1007/s11596-010-0644-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Indexed: 11/25/2022]
Abstract
This study examined the effect of ischemia-reperfusion injury on the expression of Pim-3 gene in myocardial tissues and their underlying mechanism. Rat models of myocardial ischemia-reperfusion injury were established by ligating the left anterior descending coronary artery of the rats. A total of 30 SD male adult rats were randomly divided into 5 groups: group A (sham operation, n=6); group B (in which the rats were subjected to 15 min of ischemia by ligation of the left anterior descending coronary artery, n=6); group C (in which the rats received 30 min of ischemia, n=6), group D and group E (in which the left anterior descending coronary artery of the rats were ligated for 30 min and then reperfused for 30 min or 120 min, n=6 in each). The left ventricular tissues were removed immediately after the ischemia-reperfusion injury. Neonatal cardiomyocytes were cultured and treated with different concentrations of H(2)O(2) (0, 5, 10, 20 μmol/L) or tumor necrosis factor-α (TNF-α, 0, 1, 5, 10 ng/mL). The mRNA and protein expression of Pim-3 gene was determined by using RT-PCR, western blotting and immunohistochemistry. Additionally, neonatal cardiomyocytes were transfected with Pim-3 siRNA, and induced to develop apoptosis by using H(2)O(2). The results showed that normal myocardial tissues expressed a quantity of Pim-3 gene mRNA and protein. Ischemia-reperfusion injury could up-regulate the mRNA and protein expression of Pim-3 gene in myocardial tissues. Furthermore, H(2)O(2) but not TNF-α up-regulated the Pim-3 gene expression in cultured cardiomyocytes. And Pim-3 silencing failed to strengthen the H(2)O(2)-inducing apoptosis in cardiomyocytes. It was concluded that ischemia-reperfusion injury up-regulated the Pim-3 gene expression through oxidative stress signaling pathway in myocardial tissues.
Collapse
Affiliation(s)
- Libing Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | | | | | | | | | | |
Collapse
|
38
|
Zheng Q, Schaefer AM, Nonet ML. Regulation of C. elegans presynaptic differentiation and neurite branching via a novel signaling pathway initiated by SAM-10. Development 2010; 138:87-96. [PMID: 21115607 DOI: 10.1242/dev.055350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Little is known about transcriptional control of neurite branching or presynaptic differentiation, events that occur relatively late in neuronal development. Using the Caenorhabditis elegans mechanosensory circuit as an in vivo model, we show that SAM-10, an ortholog of mammalian single-stranded DNA-binding protein (SSDP), functions cell-autonomously in the nucleus to regulate synaptic differentiation, as well as positioning of, a single neurite branch. PLM mechanosensory neurons in sam-10 mutants exhibit abnormal placement of the neurite branch point, and defective synaptogenesis, characterized by an overextended synaptic varicosity, underdeveloped synaptic morphology and disrupted colocalization of active zone and synaptic vesicles. SAM-10 functions coordinately with Lim domain-binding protein 1 (LDB-1), demonstrated by our observations that: (1) mutations in either gene show similar defects in PLM neurons; and (2) LDB-1 is required for SAM-10 nuclear localization. SAM-10 regulates PLM synaptic differentiation by suppressing transcription of prk-2, which encodes an ortholog of the mammalian Pim kinase family. PRK-2-mediated activities of SAM-10 are specifically involved in PLM synaptic differentiation, but not other sam-10 phenotypes such as neurite branching. Thus, these data reveal a novel transcriptional signaling pathway that regulates neuronal specification of neurite branching and presynaptic differentiation.
Collapse
Affiliation(s)
- Qun Zheng
- Department of Anatomy and Neurobiology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | | | | |
Collapse
|
39
|
Kim K, Kim JH, Youn BU, Jin HM, Kim N. Pim-1 Regulates RANKL-Induced Osteoclastogenesis via NF-κB Activation and NFATc1 Induction. THE JOURNAL OF IMMUNOLOGY 2010; 185:7460-6. [DOI: 10.4049/jimmunol.1000885] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Zhao H, Liu YQ, Li HX. Significance of Pim-3 expression in esophageal squamous cell carcinoma. Shijie Huaren Xiaohua Zazhi 2010; 18:2976-2980. [DOI: 10.11569/wcjd.v18.i28.2976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the significance of Pim-3 mRNA and protein expression in esophageal squamous cell carcinoma (ESCC).
METHODS: The expression of Pim-3 mRNA and protein in 45 normal esophageal mucosal specimens, 45 ESCC specimens and 22 tumor-adjacent mucosal specimens was detected by in situ hybridization and immunohistochemistry, respectively.
RESULTS: The positive rates of Pim-3 mRNA expression were 0.00% (0/45), 22.73% (5/22) and 82.22% (37/45) in normal esophageal mucosa, tumor-adjacent mucosa and ESCC tissue, respectively, with a significant difference among the three groups (χ2 = 67.450, P < 0.01). Similar results were obtained for Pim-3 protein expression: the positive rates of Pim-3 protein expression were 0.00% (0/45), 18.18% (4/22) and 75.56% (34/45) in normal esophageal mucosa, tumor-adjacent mucosa and ESCC tissue, respectively, with a significant difference among the three groups (χ2 = 60.326, P < 0.01). The expression of Pim-3 mRNA and protein was significantly associated with TNM stage and lymph node metastasis in ESCC (both P < 0.05). A positive correlation was noted between the expression of Pim-3 mRNA and protein (γp = 0.547, P < 0.01).
CONCLUSION: High expression of Pim-3 mRNA and protein may be closely related to the occurrence and development of ESCC.
Collapse
|
41
|
Wang C, Liu YQ, Feng TP, Zhao JZ, Li HX. Pim-3 knockdown suppresses cell proliferation but promotes apoptosis in esophageal squamous cell carcinoma cell line EC9706. Shijie Huaren Xiaohua Zazhi 2010; 18:2846-2851. [DOI: 10.11569/wcjd.v18.i27.2846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of down-regulation of Pim-3 expression on cell proliferation and apoptosis in esophageal squamous cell carcinoma (ESCC) cell line EC9706 and to explore possible molecular mechanisms involved.
METHODS: Small interfering RNA (siRNA) against the Pim-3 gene was transfected into EC9706 cells using LipofectamineTM 2000. After transfection, Pim-3 mRNA and protein expression was detected by RT-PCR and Western blot, respectively; cell proliferation was determined using Cell Counting Kit-8 (CCK-8); and apoptosis was measured by flow cytometry. Furthermore, the mRNA and protein expression of P21, Bcl-2 and Bax was detected by RT-PCR and Western blot, respectively.
RESULTS: Pim-3 mRNA and protein expression was significantly down-regulated after transfection with Pim-3 siRNA (both P < 0.05). Compared with untransfected cells and cells transfected with control siRNA, the proliferation of EC9706 cells was significantly restrained after transfection with Pim-3 siRNA for 24, 48, 72 and 96 h (96 h: 0.878 ± 0.061 vs 2.254 ± 0.062 and 2.219 ± 0.064, both P < 0.05), and the early apoptosis rate of EC9706 cells transfected with Pim-3 siRNA was significantly increased (19.70% ± 1.46% vs 5.35% ± 0.80% and 5.50% ± 0.61%, F = 195.692, P = 0.000). In addition, Pim-3 knockdown up-regulated P21 expression but down-regulated Bax and Bcl-2 expression in EC9706 cells (all P < 0.05).
CONCLUSION: Pim-3 plays an important role in the proliferation and apoptosis of EC9706 cells.
Collapse
|
42
|
Vlacich G, Nawijn MC, Webb GC, Steiner DF. Pim3 negatively regulates glucose-stimulated insulin secretion. Islets 2010; 2:308-17. [PMID: 21099329 PMCID: PMC3025049 DOI: 10.4161/isl.2.5.13058] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/14/2010] [Accepted: 07/16/2010] [Indexed: 01/29/2023] Open
Abstract
Pancreatic β-cell response to glucose stimulation is governed by tightly regulated signaling pathways which have not been fully characterized. A screen for novel signaling intermediates identified Pim3 as a glucose-responsive gene in the β cell, and here, we characterize its role in the regulation of β-cell function. Pim3 expression in the β-cell was first observed through microarray analysis on glucose-stimulated murine insulinoma (MIN6) cells where expression was strongly and transiently induced. In the pancreas, Pim3 expression exhibited similar dynamics and was restricted to the β cell. Perturbation of Pim3 function resulted in enhanced glucose-stimulated insulin secretion, both in MIN6 cells and in isolated islets from Pim3-/- mice, where the augmentation was specifically seen in the second phase of secretion. Consequently, Pim3-/- mice displayed an increased glucose tolerance in vivo. Interestingly, Pim3-/- mice also exhibited increased insulin sensitivity. Glucose stimulation of isolated Pim3-/- islets resulted in increased phosphorylation of ERK1/2, a kinase involved in regulating β-cell response to glucose. Pim3 was also found to physically interact with SOCS6 and SOCS6 levels were strongly reduced in Pim3-/- islets. Overexpression of SOCS6 inhibited glucose-induced ERK1/2 activation, strongly suggesting that Pim3 regulates ERK1/2 activity through SOCS6. These data reveal that Pim3 is a novel glucose-responsive gene in the β cell that negatively regulates insulin secretion by inhibiting the activation of ERK1/2, and through its effect on insulin sensitivity, has potentially a more global function in glucose homeostasis.
Collapse
Affiliation(s)
- Gregory Vlacich
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
43
|
Qureshi IA, Mehler MF. Epigenetic mechanisms underlying human epileptic disorders and the process of epileptogenesis. Neurobiol Dis 2010; 39:53-60. [PMID: 20188170 PMCID: PMC2874104 DOI: 10.1016/j.nbd.2010.02.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 02/10/2010] [Accepted: 02/13/2010] [Indexed: 10/19/2022] Open
Abstract
The rapidly emerging science of epigenetics and epigenomic medicine promises to reveal novel insights into the susceptibility to and the onset and progression of epileptic disorders. Epigenetic regulatory mechanisms are now implicated in orchestrating aspects of neural development (e.g., cell fate specification and maturation), homeostasis and stress responses (e.g., immediate early gene transcription), and neural network function (e.g., excitation-inhibition coupling and activity-dependent plasticity). These same neurobiological processes are responsible for determining the heterogeneous features of complex epileptic disease states. Thus, we highlight recent evidence that is beginning to elucidate the specific roles played by epigenetic mechanisms, including DNA methylation, histone code modifications and chromatin remodeling, noncoding RNAs and RNA editing, in human epilepsy syndromes and in the process of epileptogenesis. The highly integrated layers of the epigenome are responsible for the cell type specific and exquisitely environmentally responsive deployment of genes and functional gene networks that underlie the molecular pathophysiology of epilepsy and its associated comorbidities, including but not limited to neurotransmitter receptors (e.g., GluR2, GLRA2, and GLRA3), growth factors (e.g., BDNF), extracellular matrix proteins (e.g., RELN), and diverse transcriptional regulators (e.g., CREB, c-fos, and c-jun). These important observations suggest that future epigenetic studies are necessary to better understand, classify, prevent, and treat epileptic disorders.
Collapse
Affiliation(s)
- Irfan A. Qureshi
- Rosyln and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Mark F. Mehler
- Rosyln and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
44
|
Wu Y, Wang YY, Nakamoto Y, Li YY, Baba T, Kaneko S, Fujii C, Mukaida N. Accelerated hepatocellular carcinoma development in mice expressing the Pim-3 transgene selectively in the liver. Oncogene 2010; 29:2228-2237. [PMID: 20101231 DOI: 10.1038/onc.2009.504] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 10/25/2009] [Accepted: 12/07/2009] [Indexed: 02/05/2023]
Abstract
Pim-3, a proto-oncogene with serine/threonine kinase activity, was enhanced in hepatocellular carcinoma (HCC) tissues. To address the roles of Pim-3 in HCC development, we prepared transgenic mice that express human Pim-3 selectively in liver. The mice were born at a Mendelian ratio, were fertile and did not exhibit any apparent pathological changes in the liver until 1 year after birth. Pim-3-transgenic mouse-derived hepatocytes exhibited accelerated cell cycle progression. The administration of a potent hepatocarcinogen, diethylnitrosamine (DEN), induced accelerated proliferation of liver cells in Pim-3 transgenic mice in the early phase, compared with that observed for wild-type mice. Treatment with DEN induced lipid droplet accumulation with increased proliferating cell numbers 6 months after the treatment. Eventually, wild-type mice developed HCC with a frequency of 40% until 10 month after the treatment. Lipid accumulation was accelerated in Pim-3 transgenic mice with higher proliferating cell numbers, compared with that observed for wild-type mice. Pim-3 transgenic mice developed HCC with a higher incidence (80%) and a heavier burden, together with enhanced intratumoral CD31-positive vascular areas, compared with that observed for wild-type mice. These observations indicate that Pim-3 alone cannot cause, but can accelerate HCC development when induced by a hepatocarcinogen, such as DEN.
Collapse
Affiliation(s)
- Y Wu
- Department of Hematology and Hematology research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Brault L, Gasser C, Bracher F, Huber K, Knapp S, Schwaller J. PIM serine/threonine kinases in the pathogenesis and therapy of hematologic malignancies and solid cancers. Haematologica 2010; 95:1004-15. [PMID: 20145274 DOI: 10.3324/haematol.2009.017079] [Citation(s) in RCA: 301] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The identification as cooperating targets of Proviral Integrations of Moloney virus in murine lymphomas suggested early on that PIM serine/threonine kinases play an important role in cancer biology. Whereas elevated levels of PIM1 and PIM2 were mostly found in hematologic malignancies and prostate cancer, increased PIM3 expression was observed in different solid tumors. PIM kinases are constitutively active and their activity supports in vitro and in vivo tumor cell growth and survival through modification of an increasing number of common as well as isoform-specific substrates including several cell cycle regulators and apoptosis mediators. PIM1 but not PIM2 seems also to mediate homing and migration of normal and malignant hematopoietic cells by regulating chemokine receptor surface expression. Knockdown experiments by RNA interference or dominant-negative acting mutants suggested that PIM kinases are important for maintenance of a transformed phenotype and therefore potential therapeutic targets. Determination of the protein structure facilitated identification of an increasing number of potent small molecule PIM kinase inhibitors with in vitro and in vivo anticancer activity. Ongoing efforts aim to identify isoform-specific PIM inhibitors that would not only help to dissect the kinase function but hopefully also provide targeted therapeutics. Here, we summarize the current knowledge about the role of PIM serine/threonine kinases for the pathogenesis and therapy of hematologic malignancies and solid cancers, and we highlight structural principles and recent progress on small molecule PIM kinase inhibitors that are on their way into first clinical trials.
Collapse
Affiliation(s)
- Laurent Brault
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
46
|
Blackiston DJ, McLaughlin KA, Levin M. Bioelectric controls of cell proliferation: ion channels, membrane voltage and the cell cycle. Cell Cycle 2009; 8:3527-36. [PMID: 19823012 DOI: 10.4161/cc.8.21.9888] [Citation(s) in RCA: 312] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
All cells possess long-term, steady-state voltage gradients across the plasma membrane. These transmembrane potentials arise from the combined activity of numerous ion channels, pumps and gap junction complexes. Increasing data from molecular physiology now reveal that the role of changes in membrane voltage controls, and is in turn controlled by, progression through the cell cycle. We review recent functional data on the regulation of mitosis by bioelectric signals, and the function of membrane voltage and specific potassium, sodium and chloride ion channels in the proliferation of embryonic, somatic and neoplastic cells. Its unique properties place this powerful, well-conserved, but still poorly-understood signaling system at the center of the coordinated cellular interactions required for complex pattern formation. Moreover, disregulation of ion channel expression and function is increasingly observed to be not only a useful marker but likely a functional element in oncogenesis. New advances in genomics and the development of in vivo biophysical techniques suggest exciting opportunities for molecular medicine, bioengineering and regenerative approaches to human health.
Collapse
Affiliation(s)
- Douglas J Blackiston
- Biology Department, and Center for Regenerative and Developmental Biology, Tufts University, Medford, MA, USA
| | | | | |
Collapse
|
47
|
Liu D, He M, Yi B, Guo WH, Que AL, Zhang JX. Pim-3 protects against cardiomyocyte apoptosis in anoxia/reoxygenation injury via p38-mediated signal pathway. Int J Biochem Cell Biol 2009; 41:2315-22. [PMID: 19505587 DOI: 10.1016/j.biocel.2009.05.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 05/12/2009] [Accepted: 05/28/2009] [Indexed: 01/18/2023]
Abstract
Although anoxic preconditioning (APC) in the myocardium has been investigated for many years, its physiological mechanism is still not completely understood. Increasing evidence indicates that transiently increased resistance to ischemic damage following APC is dependent on de novo proteins synthesis. However, the key effector pathway(s) associated with APC still remains unclear. The proto-oncogene Pim kinase belongs to a serine/threoine protein kinase family, consists of Pim-1, Pim-2 and Pim-3 and has been implicated in stimulating cell growth and inhibiting cell apoptosis. Therefore we assumed that Pim-3 expression might be aberrantly induced in cardiomyocytes that were subjected to anoxia/reoxygenation (A/R) injury and that Pim-3 might also contribute to cardio-protection after APC. To address this hypothesis, we cloned a Pim-3 expression vector, transfected it into rat cardiomyocytes, and examined Pim-3 expression in rat cardiomyocytes that were subjected to A/R injury. Moreover, we studied the role of three major MAPK pathways, e.g. p38 MAPK, JNK, and ERK1/2, in order to evaluate the molecular mechanism underlying Pim-3 up-regulation and A/R induced cardiomyocyte injury. Our experiments showed that APC induced an up-regulation of Pim-3 and the transfection of Pim-3 gene into the cardiomyocytes attenuated A/R injury. The inhibition of p38 MAPK by SB203580 abolished both the Pim-3 up-regulation and the cardio-protection provided by APC. Overall, these results suggest that APC could act to protect the heart from A/R injury with cooperation from the proto-oncogene Pim-3; in addition, it up-regulates Pim-3 expression through a p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Dan Liu
- Jiangxi Provincial Key Laboratory of Molecular Medicine, Second Affiliated Hospital, Nanchang University, 1 Minde Road, Nanchang 330006, Jiangxi Province, PR China
| | | | | | | | | | | |
Collapse
|
48
|
Gong J, Wang J, Ren K, Liu C, Li B, Shi Y. Serine/threonine kinase Pim-2 promotes liver tumorigenesis induction through mediating survival and preventing apoptosis of liver cell. J Surg Res 2009; 153:17-22. [PMID: 18675992 DOI: 10.1016/j.jss.2008.03.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Revised: 03/04/2008] [Accepted: 03/25/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND It has been proven that serine/threonine kinase pim-2 mediates cell survival and prevents apoptosis in hematopoietic system tumors and lymphomas, but its role in solid organ tumor induction is still unclear. In this study, we investigated its effects and underlying mechanisms in tumorigenesis of hepatocellular carcinoma. METHODS We first examined the pim-2 gene expression and its protein levels in human hepatocellular carcinoma, paired noncancerous liver, and normal liver tissues. Then, we cultured human liver cancer cells and immortalized liver cells to examine the effects of pim-2 gene on the cell viability, growth, and apoptosis in different culture conditions. For further investigation of the molecular events in the pim-2 signal pathway, we also explored pim-2 kinase activity on phosphorylation of the two downstream signal mediators: 4E-BP1 and Bad. RESULTS Pim-2 gene and protein were notably expressed in human liver cancer tissues and HepG2 cells. The ectopic pim-2 overexpressing L02 cells were able to survive in interleukin-3 (IL-3)-deprived circumstance but not in glucose-free medium. Compared with HepG2 cells, pim-2 knock-down HepG2 cells lost survival ability in IL-3 starvation medium. In pim-2-expressing cells, both the total protein expressions of 4E-BP1 and Bad were kept stable; however, their phosphorylated patterns were notably increased. CONCLUSIONS Our results indicate that pim-2 acts as a pro-survival kinase to inhibit apoptosis and keep liver cell survival in IL-3-deprived medium. Pim-2 might participate in the tumorigenesis of hepatocellular carcinoma induction through its downstream molecules 4E-BP1 and Bad.
Collapse
Affiliation(s)
- Jianping Gong
- Department of General Surgery, 2nd College of Clinical Medicine, 2nd Affiliated Hospital of Chongqing University of Medical Science, Chongqing, China
| | | | | | | | | | | |
Collapse
|
49
|
Olla S, Manetti F, Crespan E, Maga G, Angelucci A, Schenone S, Bologna M, Botta M. Indolyl-pyrrolone as a new scaffold for Pim1 inhibitors. Bioorg Med Chem Lett 2009; 19:1512-6. [PMID: 19179076 DOI: 10.1016/j.bmcl.2009.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 12/22/2008] [Accepted: 01/06/2009] [Indexed: 11/19/2022]
Abstract
Pim1 belongs to a family of serine/threonine kinases, which is involved in the control of cell growth, differentiation, and apoptosis. Pim1 plays a pivotal role in cytokine signaling and is implicated in the development of a large number of tumors, representing a very attractive target for anticancer therapy. In this work, we applied a virtual screening protocol aimed at identifying small molecules able to inhibit Pim1 activity. The search of novel inhibitors was performed through a structure-based molecular modeling approach, taking advantage of the availability of the three-dimensional crystal structure of inhibitors bound to Pim1. Starting from the knowledge of protein-ligand complexes, the software LigandScout was used to generate pharmacophoric models, in turn used as queries to perform a virtual screening of databases, followed by docking experiments. As a result, a restricted set of candidates for biological testing was identified. Finally, among the six compounds selected as potential inhibitors of Pim1, two candidates endowed with a significant activity against Pim1 emerged. Interestingly, one of these compounds has a chemical scaffold different from inhibitors previously identified.
Collapse
Affiliation(s)
- Stefania Olla
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hsi ED, Jung SH, Lai R, Johnson JL, Cook JR, Jones D, Devos S, Cheson BD, Damon LE, Said J. Ki67 and PIM1 expression predict outcome in mantle cell lymphoma treated with high dose therapy, stem cell transplantation and rituximab: a Cancer and Leukemia Group B 59909 correlative science study. Leuk Lymphoma 2008; 49:2081-90. [PMID: 19021050 PMCID: PMC4011712 DOI: 10.1080/10428190802419640] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The proliferation index in mantle cell lymphoma (MCL) has not been validated in the context of aggressive therapy regimens in the rituximab era. We assessed Ki67 and PIM1 (a cell cycle-related gene upregulated in blastoid MCL) expression by immunohistochemistry in a phase II study Cancer and Leukemia Group B 59909 of aggressive chemotherapy and rituximab followed by autologous stem cell transplantation plus rituximab in untreated MCL patients <70 years of age. As a continuous variable or using a cutoff of 35%, higher image analysis (IA Ki67, n = 52) was associated with shorter progression free survival (PFS) (P < or = 0.030) and event free survival (EFS) (P < or = 0.017). PIM1 expression (n = 50) was associated with PFS (P = 0.033) and EFS (P = 0.043). Bivariate Cox models showed IA Ki67 and PIM1 were independent of clinical factors. High Ki67 (>35%) is an important independent prognostic marker in aggressively treated MCL in the rituximab era. PIM1 expression predicts poor outcome and, given its potential role as a therapeutic target, deserves further study.
Collapse
Affiliation(s)
- Eric D Hsi
- Department of Clinical Pathology, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|