1
|
Wang W, Jiang Q, Tao J, Zhang Z, Liu G, Qiu B, Hu Q, Zhang Y, Xie C, Song J, Jiang G, Zhong H, Zou Y, Li J, Lv S. A structure-based approach to discover a potential isomerase Pin1 inhibitor for cancer therapy using computational simulation and biological studies. Comput Biol Chem 2025; 114:108290. [PMID: 39586226 DOI: 10.1016/j.compbiolchem.2024.108290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 10/03/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Peptidyl-prolyl cis/trans isomerase Pin1 occupies a prominent role in preventing the development of certain malignant tumors. Pin1 is considered a target for the treatment of related malignant tumors, so the identification of novel Pin1 inhibitors is particularly urgent. In this study, we preliminarily predicted eight candidates from FDA-approved drug database as the potential Pin1 inhibitors through virtual screening combined with empirical screening. Therefore, we selected these eight candidates and tested their binding affinity and inhibitory activity against Pin1 using fluorescence titration and PPIase activity assays, respectively. Subsequently, we found that four FDA-approved drugs showed good binding affinities and inhibition effects. In addition, we also observed that bexarotene can reduce cell viability in a dose-dependent and time-dependent manner and induce apoptosis. Finally, we inferred that residues K63, R68 and R69 are important in the binding process between bexarotene and Pin1. All in all, repurposing of FDA-approved drugs to inhibit Pin1 may provide a promising insight into the identification and development of new treatments for certain malignant tumors.
Collapse
Affiliation(s)
- Wang Wang
- School of Basic Medicine, Nanchang Medical College, Nanchang 330006, PR China; Key Laboratory of Pharmacodynamics and Quality Evaluation on ant-inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang 330006, PR China; Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang 330006, PR China
| | - Qizhou Jiang
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Jiaxin Tao
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Zhenxian Zhang
- School of Laboratory Medicine, Nanchang Medical College, Nanchang 330006, PR China
| | - GuoPing Liu
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Binxuan Qiu
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Qingyang Hu
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Yuxi Zhang
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Chao Xie
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Jiawen Song
- School of Laboratory Medicine, Nanchang Medical College, Nanchang 330006, PR China
| | - GuoZhen Jiang
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Hui Zhong
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Yanling Zou
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Jiaqi Li
- School of Pharmacy, Nanchang Medical College, Nanchang 330006, PR China
| | - Shaoli Lv
- School of Basic Medicine, Nanchang Medical College, Nanchang 330006, PR China.
| |
Collapse
|
2
|
Chellappan MC, Vasu S, Mahadevan S, Kathiravan MK, Jayaraman S, Naik S. Beneficial Effects of PIN1 Inhibition on Diabetes Mellitus: A Concise Review. Endocr Metab Immune Disord Drug Targets 2025; 25:2-7. [PMID: 38693739 DOI: 10.2174/0118715303297663240307060019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 05/03/2024]
Abstract
Type 2 diabetes mellitus is a long-term medical illness in which the body either becomes resistant to insulin or fails to produce it sufficiently. Mostly, combinatorial therapy is required to control blood glucose levels. However, combinatorial therapy has detrimental side effects. The prevalence of the cases and subsequent increases in medical costs of the same intimidate human health globally. While there have been a lot of studies focused on developing diabetic regimens that work to lower blood glucose levels, their effectiveness is short-lived because of unfavorable side effects, such as weight gain and hypoglycemia. In recent years, the PIN1 (protein interacting with NIMA) enzyme has attracted the attention of researchers. Previous studies suggested that PIN1 may act on the various substrates that are involved in the progression of T2DM and also help in the management of diabetes-related disorders. Thus, the focus of the current review is to examine the correlation between PIN1, T2DM and its related disorders and explore the possibility of developing novel therapeutic targets through PIN1 inhibition.
Collapse
Affiliation(s)
- Meeramol C Chellappan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| | - Soumya Vasu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| | - Shriraam Mahadevan
- Department of Endocrinology, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| | | | - Saravanan Jayaraman
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, India
| | - Soniya Naik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| |
Collapse
|
3
|
Kim HS, Jung H, Park YH, Heo SH, Kim S, Moon M. Skin-brain axis in Alzheimer's disease - Pathologic, diagnostic, and therapeutic implications: A Hypothetical Review. Aging Dis 2024; 16:901-916. [PMID: 38739932 PMCID: PMC11964427 DOI: 10.14336/ad.2024.0406] [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: 01/29/2024] [Accepted: 04/06/2024] [Indexed: 05/16/2024] Open
Abstract
The dynamic interaction between the brain and the skin is termed the 'skin-brain axis.' Changes in the skin not only reflect conditions in the brain but also exert direct and indirect effects on the brain. Interestingly, the connection between the skin and brain is crucial for understanding aging and neurodegenerative diseases. Several studies have shown an association between Alzheimer's disease (AD) and various skin disorders, such as psoriasis, bullous pemphigoid, and skin cancer. Previous studies have shown a significantly increased risk of new-onset AD in patients with psoriasis. In contrast, skin cancer may reduce the risk of developing AD. Accumulating evidence suggests an interaction between skin disease and AD; however, AD-associated pathological changes mediated by the skin-brain axis are not yet clearly defined. While some studies have reported on the diagnostic implications of the skin-brain axis in AD, few have discussed its potential therapeutic applications. In this review, we address the pathological changes mediated by the skin-brain axis in AD. Furthermore, we summarize (1) the diagnostic implications elucidated through the role of the skin-brain axis in AD and (2) the therapeutic implications for AD based on the skin-brain axis. Our review suggests that a potential therapeutic approach targeting the skin-brain axis will enable significant advances in the treatment of AD.
Collapse
Affiliation(s)
- Hyeon soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Su-Hak Heo
- Department of Medicinal Bioscience, Konkuk University (Glocal Campus), Chungcheongbuk-do 27478, Korea.
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
- Research Institute for Dementia Science, Konyang University, Daejeon 35365, Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
- Research Institute for Dementia Science, Konyang University, Daejeon 35365, Korea.
| |
Collapse
|
4
|
Bai Y, Yuan Z, Yuan S, He Z. Recent advances of Pin1 inhibitors as potential anticancer agents. Bioorg Chem 2024; 144:107171. [PMID: 38325131 DOI: 10.1016/j.bioorg.2024.107171] [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: 12/09/2023] [Revised: 01/12/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Pin1 (proline isomerase peptidyl-prolyl isomerase NIMA-interacting-1), as a member of PPIase family, catalyzes cis-trans isomerization of pThr/Ser-Pro amide bonds of its substrate proteins, further regulating cell proliferation, division, apoptosis, and transformation. Pin1 is overexpressed in various cancers and is positively correlated with tumor initiation and progression. Pin1 inhibition can effectively reduce tumor growth and cancer stem cell expansion, block metastatic spread, and restore chemosensitivity, suggesting that targeting Pin1 may be an effective strategy for cancer treatment. Considering the promising therapeutic effects of Pin1 inhibitors on cancers, we herein are intended to comprehensively summarize the reported Pin1 inhibitors, mainly highlighting their structures, biological functions and binding modes, in hope of providing a reference for the future drug discovery.
Collapse
Affiliation(s)
- Yiru Bai
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Ziqiao Yuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China.
| | - Zhangxu He
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China.
| |
Collapse
|
5
|
Shrestha P, Kim G, Kang H, Bhattarai PY, Choi HS. The PIN1-YTHDF1 axis promotes breast tumorigenesis via the m 6A-dependent stabilization of AURKA mRNA. Arch Pharm Res 2024; 47:66-81. [PMID: 38147203 DOI: 10.1007/s12272-023-01480-z] [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: 08/17/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
The post-transcriptional processing of N6-methyladenosine (m6A)-modified mRNA by YTH domain-containing family protein 1 (YTHDF1) plays a crucial role in the regulation of gene expression. Although YTHDF1 expression is frequently upregulated in breast cancer, the regulatory mechanisms for this remain unclear. In this study, we examined the role of peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1) in regulating YTHDF1 stability in breast cancer cells. The WW domain of PIN1 interacted with YTHDF1 in a phosphorylation-dependent manner. Additionally, PIN1 overexpression increased YTHDF1 stability by preventing ubiquitin-dependent proteasomal degradation. Furthermore, using the MS2-tagged RNA pull-down assay, we identified Aurora kinase A (AURKA) mRNA as a bona fide substrate of YTHDF1. PIN1-mediated YTHDF1 stabilization increased the stability of AURKA mRNA in an m6A-dependent manner. Furthermore, YTHDF1 knockout reduced AURKA protein expression levels, resulting in anticancer effects in breast cancer cells, including decreased cell proliferation, cell cycle arrest at the G0/G1 phase, apoptotic cell death, and decreased spheroid formation. The anticancer effects induced by YTHDF1 knockout were reversed by AURKA overexpression. Similarly, the knockout of PIN1 produced comparable anticancer effects to those observed in YTHDF1-knockout cells, and these effects were reversed upon overexpression of YTHDF1. In conclusion, the findings of our study suggest that increased YTHDF1 stability induced by PIN1 promotes breast tumorigenesis via the stabilization of AURKA mRNA. Targeting the PIN1/YTHDF1 axis may represent a novel therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
| | - Garam Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hyelim Kang
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | | | - Hong Seok Choi
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
6
|
Naito M, Ikeda K, Aoyama S, Kanamoto M, Akasaka Y, Kido Y, Nakanishi M, Kanna M, Yamamotoya T, Matsubara A, Hinata N, Asano T, Nakatsu Y. Par14 interacts with the androgen receptor, augmenting both its transcriptional activity and prostate cancer proliferation. Cancer Med 2022; 12:8464-8475. [PMID: 36583514 PMCID: PMC10134346 DOI: 10.1002/cam4.5587] [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: 06/20/2022] [Revised: 11/14/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a major cause of cancer morbidity and mortality for men globally, and androgen signaling clearly drives its onset and progression. Androgen receptor (AR) regulation is complex and remains elusive, despite several studies tackling these issues. Therefore, elucidating the mechanism(s) underlying AR regulation is a potentially promising approach to suppressing PCa. METHODS We report that Par14, one isoform of the prolyl isomerases homologous to Pin1, is a critical regulator of AR transcriptional activity and is essential for PCa cell growth. RESULTS Par14 was shown to be overexpressed in PCa, based on analyses of deposited data. Importantly, overexpression of Par14 significantly enhanced androgen-sensitive LNCap cell growth. In contrast, silencing of Par14 dramatically decreased cell growth in LNCap cells by causing cell cycle arrest. Mechanistically, silencing of the Par14 gene dramatically induced cyclin-dependent kinase inhibitor p21 at both the mRNA and the protein level through modulating the localization of p53. In addition, suppression of Par14 in LNCap cells was shown to downregulate the expressions of androgen response genes, at both the mRNA and the protein level, induced by dihydrotestosterone. Par14 was shown to directly associate with AR in nuclei via its DNA-binding domain and augment AR transcriptional activity. CONCLUSION Thus, Par14 plays a critical role in PCa progression, and its enhancing effects on AR signaling are likely to be involved in the underlying molecular mechanisms. These findings suggest Par14 to be a promising therapeutic target for PCa.
Collapse
Affiliation(s)
- Miki Naito
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan.,Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Kenichiro Ikeda
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Shunya Aoyama
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Mayu Kanamoto
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Yasuyuki Akasaka
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan.,Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Yuri Kido
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Mikako Nakanishi
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Machi Kanna
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Takeshi Yamamotoya
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Akio Matsubara
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan.,Department of Urology, Hiroshima General Hospital, Hatsukaichi, Hiroshima, Japan
| | - Nobuyuki Hinata
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Tomoichiro Asano
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Yusuke Nakatsu
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| |
Collapse
|
7
|
In silico design of novel PIN1 inhibitors by combined of 3D-QSAR, molecular docking, molecular dynamic simulation and ADMET studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
8
|
Jug-PLGA-NPs, a New Form of Juglone with Enhanced Efficiency and Reduced Toxicity on Melanoma. Chin J Integr Med 2021; 28:909-917. [PMID: 34913148 DOI: 10.1007/s11655-021-3461-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To verrify the anti-tumor efficacy and toxicity between juglone (Jug) and Jug-loaded PLGA nanoparticles (Jug-PLGA-NPs). METHODS Jug-PLGA-NPs were prepared by ultrasonic emulsification. The anti-tumor activity of Jug (2, 3, 4 µg/mL) and Jug-PLGA-NPs (Jug: 2, 3, 4 µg/mL) in vitro was measured by MTT assay and cell apoptosis analysis. The distribution, anti-tumor effect and biological safety in vivo was evaluated on A375 nude mice. RESULTS With the advantage of good penetration and targeting properties, Jug-PLGA-NPs significantly inhibited proliferation and migration of melanoma cells both in vitro and in vivo (P<0.05 or P<0.01) with acceptable biocompatibility. CONCLUSIONS Jug can inhibit the growth of melanoma but is highly toxic. With the advantage of sustained release, tumor targeting, anti-tumor activity and acceptable biological safety, Jug-PLGA-NPs provide a new pharmaceutical form for future application of Jug.
Collapse
|
9
|
Zabłocka A, Kazana W, Sochocka M, Stańczykiewicz B, Janusz M, Leszek J, Orzechowska B. Inverse Correlation Between Alzheimer's Disease and Cancer: Short Overview. Mol Neurobiol 2021; 58:6335-6349. [PMID: 34523079 PMCID: PMC8639554 DOI: 10.1007/s12035-021-02544-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022]
Abstract
The negative association between Alzheimer's disease (AD) and cancer suggests that susceptibility to one disease may protect against the other. When biological mechanisms of AD and cancer and relationship between them are understood, the unsolved problem of both diseases which still touches the growing human population could be overcome. Actual information about biological mechanisms and common risk factors such as chronic inflammation, age-related metabolic deregulation, and family history is presented here. Common signaling pathways, e.g., p53, Wnt, role of Pin1, and microRNA, are discussed as well. Much attention is also paid to the potential impact of chronic viral, bacterial, and fungal infections that are responsible for the inflammatory pathway in AD and also play a key role to cancer development. New data about common mechanisms in etiopathology of cancer and neurological diseases suggests new therapeutic strategies. Among them, the use of nilotinib, tyrosine kinase inhibitor, protein kinase C, and bexarotene is the most promising.
Collapse
Affiliation(s)
- Agnieszka Zabłocka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland.
| | - Wioletta Kazana
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| | - Marta Sochocka
- Laboratory of Virology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| | - Bartłomiej Stańczykiewicz
- Department of Nervous System Diseases, Wroclaw Medical University, K. Bartla 5, 51-618, Wroclaw, Poland
| | - Maria Janusz
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| | - Jerzy Leszek
- Department of Psychiatry, Wroclaw Medical University, L. Pasteura 10, 50-367, Wroclaw, Poland
| | - Beata Orzechowska
- Laboratory of Virology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| |
Collapse
|
10
|
Jiang X, He Y, Shen Q, Duan L, Yuan Y, Tang L, Shi Y, Liu B, Zhai H, Shi P, Yang C, Chen Y. RETSAT Mutation Selected for Hypoxia Adaptation Inhibits Tumor Growth. Front Cell Dev Biol 2021; 9:744992. [PMID: 34805153 PMCID: PMC8601408 DOI: 10.3389/fcell.2021.744992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
Hypoxia occurs not only in natural environments including high altitude, underground burrows and deep sea, but also in human pathological conditions, such as hypoxic solid tumors. It has been well documented that hypoxia related signaling pathway is associated with a poor clinical outcome. Our group has recently identified multiple novel genes critical for solid tumor growth comparing the genome-wide convergent/parallel sequence evolution of highland mammals. Among them, a single mutation on the retinol saturase gene (RETSAT) containing amino acid switch from glutamine (Q) to arginine (R) at the position 247 was identified. Here, we demonstrate that RETSAT is mostly downregulated in multiple types of human cancers, whose lower expression correlates with worse clinical outcome. We show that higher expression of RETSAT is positively associated with immune infiltration in different human cancers. Furthermore, we identify that the promoter region of RETSAT is highly methylated, which leads to its decreased expressions in tumor tissues comparing to normal tissues. Furthermore, we show that RETSAT knockdown promotes, while its overexpression inhibits, the cell proliferation ability of mouse embryonic fibroblasts (MEFs) and B16 in vitro. In addition, the mice carrying homozygous Q247R mutation (RETSATR/R) is more resistant to xenograft tumor formation, as well as DMBA/TPA induced cutaneous keratinocyte carcinoma formation, compared to littermate wild-type (RETSATQ/Q) mice. Mechanistic study uncovers that the oncogenic factor, the prolyl isomerase (PPIase) Pin1 and its related downstream signaling pathway, were both markedly repressed in the mutant mice compared to the wild-type mice. In summary, these results suggest that interdisciplinary study between evolution and tumor biology can facilitate identification of novel molecular events essential for hypoxic solid tumor growth in the future.
Collapse
Affiliation(s)
- Xiulin Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yaomei He
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qiushuo Shen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Lincan Duan
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yixiao Yuan
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Tang
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yulin Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Baiyang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Haoqing Zhai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Cuiping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
11
|
Vashi R, Patel BM, Goyal RK. Keeping abreast about ashwagandha in breast cancer. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113759. [PMID: 33359916 DOI: 10.1016/j.jep.2020.113759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 10/29/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ashwagandha has been used as an ayurvedic medicine in the form of 'Rasayana' (as a tonic) even before 3000 BCE in India. As per Ayurveda, it has long been used traditionally for the treatment of inflammation, weakness, impotence, pulmonary tuberculosis. This plant is also beneficial in lumbago and leucorrhea in the female. In the recent past, Withania has shown its anti-cancerous activity in various experimental models. In addition, Withania also possesses many other properties such as anti-oxidant, anti-stress, adaptogenic, and regenerative which will eventually be beneficial and safe in treating cancer patients. AIM OF THE STUDY This review aims to provide experimental evidence along with a deeper insight into molecular mechanisms of Ashwagandha (Withania somnifera (L.) Dunal) through which it acts as a chemotherapeutic agent against different types of breast cancer. MATERIALS AND METHODS Literature searches with the help of electronic online databases (Elsevier, Google Scholar, Scopus, Springer Link, ScienceDirect, ResearchGate, PubMed) were carried out. The timeline for collection of data for the review article was from 2000 to 2019. The plant name was validated from The Plant List (2013). Version 1.1. Published on http://www.theplantlist.org/(accessed 21st March 2020). RESULTS Various forms of Withania somnifera were used and several in vitro, in vivo, and clinical studies were reported by researchers. They found ashwagandha to exhibit anti-apoptotic, anti-metastatic, anti-invasive and anti-inflammatory properties and gave the evidence that ashwagandha has a capability for averting and treating breast cancer. CONCLUSION Various in vitro and in vivo studies suggested Ashwagandha may possess a potential for treating breast cancer, especially ER/PR positive breast cancer and triple-negative breast cancer. A clinical trial has also been conducted in the past that suggested its potential in refining quality of life in breast cancer patients. Studies directed towards molecular pathways have helped in unravelling the key mechanisms of ashwagandha. Future research should be directed towards translational studies involving breast cancer patients. These will reinforce the ancient power of our Ayurvedic medicine.
Collapse
Affiliation(s)
- Ruju Vashi
- Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, India.
| | - Bhoomika M Patel
- Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, India.
| | - Ramesh K Goyal
- Delhi Pharmaceutical Sciences Research University, Delhi, India.
| |
Collapse
|
12
|
Maggio J, Cabrera M, Armando R, Chinestrad P, Pifano M, Menna PL, Gomez DE, Gómez DLM. Rational design of PIN1 inhibitors for cancer treatment based on conformational diversity analysis and docking based virtual screening. J Biomol Struct Dyn 2021; 40:5858-5867. [PMID: 33463409 DOI: 10.1080/07391102.2021.1874531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The parvulin PIN1 (peptidyl-prolyl cis-trans isomerase NIMA-interacting 1), is the only enzyme capable of isomerizing prolines of phospho-Serine/Threonine-Proline motifs. PIN1 binds to a subset of proteins and plays an essential role in regulating protein function post-phosphorylation control. Furthermore, the activity of PIN1 regulates the outcome of the signalling of proline-directed kinases (e.g. MAPK, CDK, or GSK3) and thus regulates cell proliferation and cell survival. For these reasons, PIN1 inhibitors are interesting since they may have therapeutic implications for cancer. Several authors have already reported that the non-structural point mutation Trp34Ala prevents PIN1 from interacting with its downstream effector proteins. In this work, we characterized PIN1 structurally, intending to explore new inhibition targets for the rational design of pharmacological activity compounds. Through a conformational diversity analysis of PIN1, we identified and characterized a highly specific druggable pocket around the residue Trp34. This pocket was used in a high-throughput docking screening of 450,000 drug-like compounds, and the top 10 were selected for re-docking studies on the previously used conformers. Finally, we evaluated the binding of each compound by thermal shift assay and found four molecules with a high affinity for PIN1 and potential inhibitory activity. Through this strategy, we achieved novel drug candidates with the ability to interfere with the phosphorylation-dependent actions of PIN1 and with potential applications in the treatment of cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Julián Maggio
- Departamento de Ciencia y Tecnología, Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Maia Cabrera
- Departamento de Ciencia y Tecnología, Laboratorio de Farmacología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Romina Armando
- Departamento de Ciencia y Tecnología, Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Patricio Chinestrad
- Departamento de Ciencia y Tecnología, Laboratorio de Farmacología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Marina Pifano
- Departamento de Ciencia y Tecnología, Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Pablo Lorenzano Menna
- Departamento de Ciencia y Tecnología, Laboratorio de Farmacología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Daniel E Gomez
- Departamento de Ciencia y Tecnología, Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Diego L Mengual Gómez
- Departamento de Ciencia y Tecnología, Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, Bernal, Argentina
| |
Collapse
|
13
|
Munakarmi S, Chand L, Shin HB, Hussein UK, Yun BS, Park HR, Jeong YJ. Anticancer effects of Poncirus fructus on hepatocellular carcinoma through regulation of apoptosis, migration, and invasion. Oncol Rep 2020; 44:2537-2546. [PMID: 33125135 PMCID: PMC7640358 DOI: 10.3892/or.2020.7790] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Poncirus fructus (PF) is a phytochemical compound extracted from the dry, immature fruits of Poncirus trifoliate. PF is traditionally used to treat gastrointestinal disorders, allergies, and inflammatory disease. In East Asia, PF is also known for its anticancer properties. There are numerous reports on the anticancer and anti-inflammatory effects of PF in a wide range of cancers and gastrointestinal diseases, respectively. However, the role of PF in inducing apoptosis and suppressing the invasiveness of hepatocellular carcinoma (HCC) remains unclear. This study investigated the ability of PF to induce apoptosis and inhibit the invasiveness and migratory ability of HCC cell lines (Hep3B and Huh7). Wound healing, Transwell migration and invasion, and colony-formation assays, as well as flow cytometry, were used to analyze cell proliferation, migration, invasion, and apoptosis. Epithelial-mesenchymal transition (EMT)-related and apoptotic proteins were assessed by western blotting. The mitochondrial membrane potential of the Hep3B and Huh7 cells was observed with tetramethylrhodamine ethyl ester. The reactive oxygen species (ROS) level was determined by dihydroethidium (DHE) staining. PF treatment significantly decreased the proliferation of Hep3B and Huh7 cells in a dose-dependent manner, reduced the mitochondrial membrane potential, increased ROS levels, decreased the protein levels of Bcl-2, and increased the protein levels of Bax and cleaved caspase-3 and 9, suggesting that PF mediated HCC apoptosis via a mitochondrial pathway. Our findings showed that PF prevented HCC cell migration and invasion by inhibiting the EMT process and downregulating MMP-2 and MMP-9 activities. The results suggest the potential anticancer effects of PF by inhibiting proliferation, inducing apoptosis, and reducing the invasion and migration of HCC cells.
Collapse
Affiliation(s)
- Suvesh Munakarmi
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Lokendra Chand
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Hyun Beak Shin
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Usama Khamis Hussein
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Chonbuk National University, Jeonju 54907, Republic of Korea
| | - Bong-Sik Yun
- Division of Biotechnology, College of Environmental and BioSource Sciences, Chonbuk National University, Jeonju 54907, Republic of Korea
| | - Hae Ryong Park
- Department of Food Science and Biotechnology, Kyungnam University, Masanhappo‑gu, Changwon‑si 631‑701, Republic of Korea
| | - Yeon Jun Jeong
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| |
Collapse
|
14
|
Yang H, Zhang P, Li J, Gao Y, Zhao L, Li J, Guo M, Zhang J, Li H, Wang F, Yuan Y. Targeting PIN-1 Attenuates GCB DLBCL Cell Proliferation Through Inhibition of PI3K/AKT Signaling. Onco Targets Ther 2020; 13:8593-8600. [PMID: 32904547 PMCID: PMC7457679 DOI: 10.2147/ott.s247429] [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: 01/28/2020] [Accepted: 07/26/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Diffuse large B cell lymphoma (DLBCL) is a highly heterogeneous type of non-Hodgkin lymphoma with many molecular subtypes that can be distinguished by gene expression profiling (GEP). However, the pathogenesis of DLBCL is still unclear. Materials and Methods The expression levels of the prolyl isomerase PIN-1 and other related proteins were determined in 73 primary DLBCL patient samples and cell lines by Western blotting (WB) and immunohistochemical (IHC) staining. Cell cycle and apoptosis were evaluated by flow cytometry. Lymphoma cell viability was detected by CCK-8 proliferation assay. Results High levels of PIN-1 expression were detected in 55% of germinal center B cell (GCB) DLBCL patient samples, whereas such abnormal expression levels were found in only 11% of non-GCB DLBCL patient samples. PIN-1 expression was positively associated with activation of the oncogenic phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway in both GCB DLBCL cell lines and primary patient samples. Depletion of PIN-1 was cytotoxic to GCB DLBCL model cell lines because it led to inhibition of the PI3K/AKT signaling pathway, revealing a GCB DLBCL subgroup that is dependent on this pathway. A PI3K inhibitor was selectively toxic to GCB DLBCL lines expressing high levels of PIN-1. Conclusion Our study used PIN-1 to identify a new subgroup of GCB DLBCL associated with the PI3K/AKT signaling pathway, and our findings reveal that inhibition of PI3K is a promising therapeutic approach for GCB DLBCL.
Collapse
Affiliation(s)
- Haijun Yang
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Ping Zhang
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Junkuo Li
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Yang Gao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100032, People's Republic of China
| | - Luyao Zhao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100032, People's Republic of China
| | - Jia Li
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Mei Guo
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Jingfang Zhang
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Haimei Li
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Fuqiang Wang
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| | - Yufen Yuan
- Department of Pathology, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, People's Republic of China
| |
Collapse
|
15
|
Nakatsu Y, Matsunaga Y, Ueda K, Yamamotoya T, Inoue Y, Inoue MK, Mizuno Y, Kushiyama A, Ono H, Fujishiro M, Ito H, Okabe T, Asano T. Development of Pin1 Inhibitors and their Potential as Therapeutic Agents. Curr Med Chem 2020; 27:3314-3329. [PMID: 30394205 DOI: 10.2174/0929867325666181105120911] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 12/26/2022]
Abstract
The prolyl isomerase Pin1 is a unique enzyme, which isomerizes the cis-trans conformation between pSer/pThr and proline and thereby regulates the function, stability and/or subcellular distribution of its target proteins. Such regulations by Pin1 are involved in numerous physiological functions as well as the pathogenic mechanisms underlying various diseases. Notably, Pin1 deficiency or inactivation is a potential cause of Alzheimer's disease, since Pin1 induces the degradation of Tau. In contrast, Pin1 overexpression is highly correlated with the degree of malignancy of cancers, as Pin1 controls a number of oncogenes and tumor suppressors. Accordingly, Pin1 inhibitors as anti-cancer drugs have been developed. Interestingly, recent intensive studies have demonstrated Pin1 to be responsible for the onset or development of nonalcoholic steatosis, obesity, atherosclerosis, lung fibrosis, heart failure and so on, all of which have been experimentally induced in Pin1 deficient mice. In this review, we discuss the possible applications of Pin1 inhibitors to a variety of diseases including malignant tumors and also introduce the recent advances in Pin1 inhibitor research, which have been reported.
Collapse
Affiliation(s)
- Yusuke Nakatsu
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Yasuka Matsunaga
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Koji Ueda
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Takeshi Yamamotoya
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Yuki Inoue
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Masa-Ki Inoue
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Yu Mizuno
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Akifumi Kushiyama
- The Division of Diabetes and Metabolism, Institute for Adult Diseases, Asahi Life Foundation, Chuo-ku, Tokyo 103-0002, Japan
| | - Hiraku Ono
- Department of Clinical Cell Biology, Chiba University Graduate School of Medicine, Chiba City, Chiba 260-8677, Japan
| | - Midori Fujishiro
- The Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan
| | - Hisanaka Ito
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Takayoshi Okabe
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| |
Collapse
|
16
|
Seo J, Park M. Molecular crosstalk between cancer and neurodegenerative diseases. Cell Mol Life Sci 2020; 77:2659-2680. [PMID: 31884567 PMCID: PMC7326806 DOI: 10.1007/s00018-019-03428-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
The progression of cancers and neurodegenerative disorders is largely defined by a set of molecular determinants that are either complementarily deregulated, or share remarkably overlapping functional pathways. A large number of such molecules have been demonstrated to be involved in the progression of both diseases. In this review, we particularly discuss our current knowledge on p53, cyclin D, cyclin E, cyclin F, Pin1 and protein phosphatase 2A, and their implications in the shared or distinct pathways that lead to cancers or neurodegenerative diseases. In addition, we focus on the inter-dependent regulation of brain cancers and neurodegeneration, mediated by intercellular communication between tumor and neuronal cells in the brain through the extracellular microenvironment. Finally, we shed light on the therapeutic perspectives for the treatment of both cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jiyeon Seo
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Mikyoung Park
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea.
- Department of Neuroscience, Korea University of Science and Technology, Daejeon, 34113, South Korea.
| |
Collapse
|
17
|
Liu K, Zheng M, Lu R, Du J, Zhao Q, Li Z, Li Y, Zhang S. The role of CDC25C in cell cycle regulation and clinical cancer therapy: a systematic review. Cancer Cell Int 2020; 20:213. [PMID: 32518522 PMCID: PMC7268735 DOI: 10.1186/s12935-020-01304-w] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 05/28/2020] [Indexed: 12/24/2022] Open
Abstract
One of the most prominent features of tumor cells is uncontrolled cell proliferation caused by an abnormal cell cycle, and the abnormal expression of cell cycle-related proteins gives tumor cells their invasive, metastatic, drug-resistance, and anti-apoptotic abilities. Recently, an increasing number of cell cycle-associated proteins have become the candidate biomarkers for early diagnosis of malignant tumors and potential targets for cancer therapies. As an important cell cycle regulatory protein, Cell Division Cycle 25C (CDC25C) participates in regulating G2/M progression and in mediating DNA damage repair. CDC25C is a cyclin of the specific phosphatase family that activates the cyclin B1/CDK1 complex in cells for entering mitosis and regulates G2/M progression and plays an important role in checkpoint protein regulation in case of DNA damage, which can ensure accurate DNA information transmission to the daughter cells. The regulation of CDC25C in the cell cycle is affected by multiple signaling pathways, such as cyclin B1/CDK1, PLK1/Aurora A, ATR/CHK1, ATM/CHK2, CHK2/ERK, Wee1/Myt1, p53/Pin1, and ASK1/JNK-/38. Recently, it has evident that changes in the expression of CDC25C are closely related to tumorigenesis and tumor development and can be used as a potential target for cancer treatment. This review summarizes the role of CDC25C phosphatase in regulating cell cycle. Based on the role of CDC25 family proteins in the development of tumors, it will become a hot target for a new generation of cancer treatments.
Collapse
Affiliation(s)
- Kai Liu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Rui Lu
- Department of Pathology, Tianjin Nankai Hospital, Tianjin, People’s Republic of China
| | - Jiaxing Du
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Qi Zhao
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Zugui Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Yuwei Li
- Departments of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| |
Collapse
|
18
|
Chen D, Wang L, Lee TH. Post-translational Modifications of the Peptidyl-Prolyl Isomerase Pin1. Front Cell Dev Biol 2020; 8:129. [PMID: 32195254 PMCID: PMC7064559 DOI: 10.3389/fcell.2020.00129] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
The peptidyl-prolyl cis/trans isomerase (PPIase) Pin1 is a unique enzyme that only binds to Ser/Thr-Pro peptide motifs after phosphorylation and regulates the conformational changes of the bond. The Pin1-catalyzed isomerization upon phosphorylation can have profound effects on substrate biological functions, including their activity, stability, assembly, and subcellular localization, affecting its role in intracellular signaling, transcription, and cell cycle progression. The functions of Pin1 are regulated by post-translational modifications (PTMs) in many biological processes, which include phosphorylation, ubiquitination, SUMOylation and oxidation. Phosphorylation of different Pin1 sites regulates Pin1 enzymatic activity, binding ability, localization, and ubiquitination by different kinases under various cellular contexts. Moreover, SUMOylation and oxidation have been shown to downregulate Pin1 activity. Although Pin1 is tightly regulated under physiological conditions, deregulation of Pin1 PTMs contributes to the development of human diseases including cancer and Alzheimer's disease (AD). Therefore, manipulating the PTMs of Pin1 may be a promising therapeutic option for treating various human diseases. In this review, we focus on the molecular mechanisms of Pin1 regulation by PTMs and the major impact of Pin1 PTMs on the progression of cancer and AD.
Collapse
Affiliation(s)
- Dongmei Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Tae Ho Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
19
|
Chen D, Zhou XZ, Lee TH. Death-Associated Protein Kinase 1 as a Promising Drug Target in Cancer and Alzheimer's Disease. Recent Pat Anticancer Drug Discov 2020; 14:144-157. [PMID: 30569876 PMCID: PMC6751350 DOI: 10.2174/1574892814666181218170257] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/23/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023]
Abstract
Background: Death-Associated Protein Kinase 1 (DAPK1) plays an important role in apopto-sis, tumor suppression and neurodegeneration including Alzheimer’s Disease (AD). Objective: This review will describe the diverse roles of DAPK1 in the development of cancer and AD, and the current status of drug development targeting DAPK1-based therapies. Methods: Reports of DAPK1 regulation, function and substrates were analyzed using genetic DAPK1 manipulation and chemical DAPK1 modulators. Results: DAPK1 expression and activity are deregulated in cancer and AD. It is down-regulated and/or inactivated by multiple mechanisms in many human cancers, and elicits a protective effect to counteract numerous death stimuli in cancer, including activation of the master regulator Pin1. Moreover, loss of DAPK1 expression has correlated strongly with tumor recurrence and metastasis, suggesting that lack of sufficient functional DAPK1 might contribute to cancer. In contrast, DAPK1 is highly expressed in the brains of most human AD patients and has been identified as one of the genetic factors affecting suscepti-bility to late-onset AD. The absence of DAPK1 promotes efficient learning and better memory in mice and prevents the development of AD by acting on many key proteins including Pin1 and its downstream tar-gets tau and APP. Recent patents show that DAPK1 modulation might be used to treat both cancer and AD. Conclusion: DAPK1 plays a critical role in diverse physiological processes and importantly, its deregula-tion is implicated in the pathogenesis of either cancer or AD. Therefore, manipulating DAPK1 activity and/or expression may be a promising therapeutic option for cancer or AD.
Collapse
Affiliation(s)
- Dongmei Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Xiao Z Zhou
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Tae H Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| |
Collapse
|
20
|
Born A, Henen MA, Vögeli B. Activity and Affinity of Pin1 Variants. MOLECULES (BASEL, SWITZERLAND) 2019; 25:molecules25010036. [PMID: 31861908 PMCID: PMC6983177 DOI: 10.3390/molecules25010036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/07/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
Pin1 is a peptidyl-prolyl isomerase responsible for isomerizing phosphorylated S/T-P motifs. Pin1 has two domains that each have a distinct ligand binding site, but only its PPIase domain has catalytic activity. Vast evidence supports interdomain allostery of Pin1, with binding of a ligand to its regulatory WW domain impacting activity in the PPIase domain. Many diverse studies have made mutations in Pin1 in order to elucidate interactions that are responsible for ligand binding, isomerase activity, and interdomain allostery. Here, we summarize these mutations and their impact on Pin1′s structure and function.
Collapse
Affiliation(s)
- Alexandra Born
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA; (A.B.); (M.A.H.)
| | - Morkos A. Henen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA; (A.B.); (M.A.H.)
- Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Beat Vögeli
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA; (A.B.); (M.A.H.)
- Correspondence: ; Tel.: +1-303-724-1627
| |
Collapse
|
21
|
Abstract
The small ubiquitin-related modification molecule (SUMO), one of the post-translational modification molecules, is involved in a variety of cellular functions where it regulates protein activity and stability, transcription, and cell cycling. Modulation of protein SUMOylation or deSUMOylation modification has been associated with regulation of carcinogenesis in breast cancer. In the dynamic processes of SUMOylation and deSUMOylation in a variety of cancers, SUMO proteases (SENPs), reverse SUMOylation by isopeptidase activity and SENPs are mostly elevated, and are related to poor patient prognosis. Although underlying mechanisms have been suggested for how SENPs participate in breast cancer tumorigenesis, such as through regulation of target protein transactivation, cancer cell survival, cell cycle, or other post-translational modification-related machinery recruitment, the effect of SENP isoform-specific inhibitors on the progression of breast cancer have not been well evaluated. This review will introduce the functions of SENP1 and SENP2 and the underlying signaling pathways in breast cancer for use in discovery of new biomarkers for diagnosis or therapeutic targets for treatment. [BMB Reports 2019; 52(2): 113-118].
Collapse
Affiliation(s)
- Kyung-Sun Heo
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
22
|
Khanal P, Yeung B, Zhao Y, Yang X. Identification of Prolyl isomerase Pin1 as a novel positive regulator of YAP/TAZ in breast cancer cells. Sci Rep 2019; 9:6394. [PMID: 31015482 PMCID: PMC6478839 DOI: 10.1038/s41598-019-42767-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/08/2019] [Indexed: 01/06/2023] Open
Abstract
The Hippo signalling pathway plays very important roles in tumorigenesis, metastasis, organ size control, and drug resistance. Although, it has been shown that the two major components of Hippo pathway, YAP and TAZ, play very crucial role in tumorigenesis and drug resistance, the exact molecular mechanisms are still unknown. Recently, we have shown that the prolyl isomerase Pin1 regulates the activity of Hippo pathway through interaction with Hippo component LATS kinase. Thus we asked if Pin1 is also able to interact with other Hippo pathway components. Therefore, in order to investigate whether Pin1 can interacts with other components of the Hippo pathway, we performed GST-pull down and co-immunoprecipitation (Co-IP) assays and have identified two Hippo components YAP and TAZ oncoproteins as novel binding partner of Pin1. We found that Pin1 interacts with YAP/TAZ in a phosphorylation-independent manner and WW domain of Pin1 is necessary for this interaction. Moreover, by using real time qRT-PCR, Cycloheximide chase, luciferase reporter, cell viability and soft agar assays, we have shown that Pin1 increases the tumorigenic and drug-resistant activity of YAP/TAZ through stabilization of YAP/TAZ at protein levels. Together, we have identified Pin1 as a novel positive regulator of YAP/TAZ in tumorigenesis and drug resistance of breast cancer cells. These findings will provide a significant contribution for targeting the Pin1-YAP/TAZ signaling for the successful treatment of tumorigenesis and drug resistance of breast and other cancers in the future.
Collapse
Affiliation(s)
- Prem Khanal
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Benjamin Yeung
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Yulei Zhao
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada.
| |
Collapse
|
23
|
Fan G, Wang L, Xu J, Jiang P, Wang W, Huang Y, Lv M, Liu S. Knockdown of the prolyl isomerase Pin1 inhibits Hep-2 cell growth, migration, and invasion by targeting the β-catenin signaling pathway. Biochem Cell Biol 2018; 96:734-741. [PMID: 29768138 DOI: 10.1139/bcb-2017-0334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence indicating that peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (Pin1) plays a decisive role in a variety of cancers. Nevertheless, its function in laryngeal squamous cell carcinoma (LSCC) has not been elaborated. The aim of this study is to determine the role of Pin1 in LSCC. Here, we established stably transfected Hep-2 cells with low expression of Pin1. Intriguingly, cell proliferation, migration, and invasion was significantly inhibited in Pin1-silenced Hep-2 cells. Similarly, knockdown of Pin1 induced apoptosis of Hep-2 cells, as evidenced by increased expression of cleaved-caspase-3, cleaved-PARP, and bax, and decreased expression of bcl2. We also demonstrated that silencing of Pin1 down-regulated β-catenin and cyclin D1 expression. Inversely, over-expression of β-catenin reversed the inhibiting effect of Pin1 silencing on Hep-2 cells. Moreover, we proved that knockdown of Pin1 inhibited tumorigenesis of Hep-2 cells in vivo. Taken together, we demonstrate that silencing of Pin1 effectively suppresses the growth of Hep-2 cells through β-catenin, indicating that Pin1 possess the potential to serve as a therapeutic target for the treatment of LSCC.
Collapse
Affiliation(s)
- Guoliang Fan
- Department of Otolaryngology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Lin Wang
- Department of Otolaryngology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Jia Xu
- Department of Otolaryngology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Ping Jiang
- Department of Pathology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Wei Wang
- Department of Otolaryngology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Ying Huang
- Department of Pathology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Minggang Lv
- Department of Otolaryngology, Harbin First Hospital, Harbin, People’s Republic of China
| | - Shaoting Liu
- Department of Otolaryngology, Harbin First Hospital, Harbin, People’s Republic of China
| |
Collapse
|
24
|
Ma T, Huang M, Li A, Zhao F, Li D, Liu D, Zhao L. Design, synthesis and biological evaluation of benzimidazole derivatives as novel human Pin1 inhibitors. Bioorg Med Chem Lett 2018; 29:1859-1863. [PMID: 31103446 DOI: 10.1016/j.bmcl.2018.11.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
In this work, a series of novel benzimidazole derivatives were designed and synthesized as Pin1 inhibitors. Protease-coupled assay was used to investigate the Pin1 inhibitory potency of all synthesized compounds. Thirteen of them showed preferable Pin1 inhibitory effects with IC50 values lower than 5 μM, and 12a, 15b, 15d and 16c exhibited the most promising Pin1 inhibitory activity at low micromolar level (0.33-1.00 μM) than the positive control compound Juglone. Flow cytometry results showed that treating PC-3 cells with 16c caused slight cycle arrest in a concentration-dependent manner. The structure-activity relationships of R1, R2, R3 and linker of the benzimidazole derivatives were analyzed in detail, which would help further exploration of new Pin1 inhibitors.
Collapse
Affiliation(s)
- Tianyi Ma
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Min Huang
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Aihua Li
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Feng Zhao
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Deyi Li
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Dan Liu
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Linxiang Zhao
- Key Laboratory of Structure-based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
25
|
Lee YM, Liou YC. Gears-In-Motion: The Interplay of WW and PPIase Domains in Pin1. Front Oncol 2018; 8:469. [PMID: 30460195 PMCID: PMC6232885 DOI: 10.3389/fonc.2018.00469] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/04/2018] [Indexed: 01/22/2023] Open
Abstract
Pin1 belongs to the family of the peptidyl-prolyl cis-trans isomerase (PPIase), which is a class of enzymes that catalyze the cis/trans isomerization of the Proline residue. Pin1 is unique and only catalyzes the phosphorylated Serine/Threonine-Proline (S/T-P) motifs of a subset of proteins. Since the discovery of Pin1 as a key protein in cell cycle regulation, it has been implicated in numerous diseases, ranging from cancer to neurodegenerative diseases. The main features of Pin1 lies in its two main domains: the WW (two conserved tryptophan) domain and the PPIase domain. Despite extensive studies trying to understand the mechanisms of Pin1 functions, how these two domains contribute to the biological roles of Pin1 in cellular signaling requires more investigations. The WW domain of Pin1 is known to have a higher affinity to its substrate than that of the PPIase domain. Yet, the WW domain seems to prefer the trans configuration of phosphorylated S/T-P motif, while the PPIase catalyzes the cis to trans isomerasion. Such contradicting information has generated much confusion as to the actual mechanism of Pin1 function. In addition, dynamic allostery has been suggested to be important for Pin1 function. Henceforth, in this review, we will be looking at the progress made in understanding the function of Pin1, and how these understandings can aid us in overcoming the diseases implicated by Pin1 such as cancer during drug development.
Collapse
Affiliation(s)
- Yew Mun Lee
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
26
|
Huang M, Li A, Zhao F, Xie X, Li K, Jing Y, Liu D, Zhao L. Design, synthesis and biological evaluation of ring A modified 11-keto-boswellic acid derivatives as Pin1 inhibitors with remarkable anti-prostate cancer activity. Bioorg Med Chem Lett 2018; 28:3187-3193. [PMID: 30153964 DOI: 10.1016/j.bmcl.2018.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 11/27/2022]
Abstract
Pin1 (Protein interaction with never in mitosis A1) is a validated molecular target for anticancer drug discovery. Herein, we reported the design, synthesis, and structure-activity relationship study of novel ring A modified AKBA (3-acetyl-11-keto-boswellic acid) derivatives as Pin1 inhibitors. Most compounds showed superior Pin1 inhibitory activities to AKBA. One of the most promising compounds, 10a, potently inhibited Pin1 with IC50 value of 0.46 μM, while it displayed excellent anti-proliferative effect against prostate cancer cells PC-3 with GI50 value of 1.82 μM. Structure-activity relationship indicated that reasonable structural modifications in ring A had significant impact on improving activity. Further mechanism research revealed that 10a decreased the level of Cyclin D1 and caused cell cycle arrest at G0/G1 phase in PC-3 cancer cells. Thus, compound 10a may serve as potential anti-prostate cancer agent for further investigation through Pin1 inhibition.
Collapse
Affiliation(s)
- Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Aihua Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Feng Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaorui Xie
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Kun Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongkui Jing
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
27
|
Cao W, Yao J, Feng S, He Y, Jiang E, Zhang R, Yang D, Gong M, Zheng X, Chen S, Sun J, Zhou L, Han M. BCR-ABL enhances the prolyl isomerase activity of Pin 1 by interacting with DAPK1 in ph + ALL. Cancer Med 2018; 7:2530-2540. [PMID: 29665256 PMCID: PMC6010889 DOI: 10.1002/cam4.1478] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/06/2018] [Accepted: 02/09/2018] [Indexed: 12/27/2022] Open
Abstract
Philadelphia chromosome (Ph)/BCR-ABL-positive (ph+ ) ALL is the most common genetic abnormality associated with ALL and has been shown to confer the worst prognosis to both children and adults. Increasing evidence has revealed that the dysregulation of prolyl isomerase Pin 1 contributes to multicancer development and progression, including ALL, although the underlying molecular mechanisms remain unclear. Here, we report that the expression of Pin 1 was enhanced in ph+ ALL patient samples and was associated positively with the expression of BCR-ABL. Genetically or pharmacologically inhibiting Pin 1 expression or activity produces potent therapeutic efficacy against ph+ ALL. We further demonstrated that BCR-ABL enhances the prolyl isomerase activity of Pin 1 by decreasing the phosphorylated level of Pin 1 at Ser 71 and interacting with DAPK1. The inhibition of BCR-ABL activity by imatinib in human ph+ ALL cells reduces the prolyl isomerase activity of Pin 1, further suggesting a key role of the newly identified BCR-ABL-Pin 1 axis in ph+ ALL progression. Thus, the combined suppression of Pin 1 and BCR-ABL proteins may be exploited as an additional target therapy for ph+ ALL.
Collapse
Affiliation(s)
- Wen‐bin Cao
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Jian‐feng Yao
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Si‐zhou Feng
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Yi He
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Er‐lie Jiang
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Rong‐li Zhang
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Dong‐lin Yang
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Ming Gong
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Xiao‐hui Zheng
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Shu‐lian Chen
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Jia‐li Sun
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Lu‐kun Zhou
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| | - Ming‐zhe Han
- Hematopoietic Stem Cell Transplantation CenterInstitute of Hematology and Blood Diseases HospitalPeking Union Medical College and Chinese Academy of Medical SciencesNo. 288 Nanjing Road300020TianjinChina
| |
Collapse
|
28
|
Lian X, Lin YM, Kozono S, Herbert MK, Li X, Yuan X, Guo J, Guo Y, Tang M, Lin J, Huang Y, Wang B, Qiu C, Tsai CY, Xie J, Gao ZJ, Wu Y, Liu H, Zhou XZ, Lu KP, Chen Y. Pin1 inhibition exerts potent activity against acute myeloid leukemia through blocking multiple cancer-driving pathways. J Hematol Oncol 2018; 11:73. [PMID: 29848341 PMCID: PMC5977460 DOI: 10.1186/s13045-018-0611-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/29/2018] [Indexed: 12/14/2022] Open
Abstract
Background The increasing genomic complexity of acute myeloid leukemia (AML), the most common form of acute leukemia, poses a major challenge to its therapy. To identify potent therapeutic targets with the ability to block multiple cancer-driving pathways is thus imperative. The unique peptidyl-prolyl cis-trans isomerase Pin1 has been reported to promote tumorigenesis through upregulation of numerous cancer-driving pathways. Although Pin1 is a key drug target for treating acute promyelocytic leukemia (APL) caused by a fusion oncogene, much less is known about the role of Pin1 in other heterogeneous leukemia. Methods The mRNA and protein levels of Pin1 were detected in samples from de novo leukemia patients and healthy controls using real-time quantitative RT-PCR (qRT-PCR) and western blot. The establishment of the lentiviral stable-expressed short hairpin RNA (shRNA) system and the tetracycline-inducible shRNA system for targeting Pin1 were used to analyze the biological function of Pin1 in AML cells. The expression of cancer-related Pin1 downstream oncoproteins in shPin1 (Pin1 knockdown) and Pin1 inhibitor all-trans retinoic acid (ATRA) treated leukemia cells were examined by western blot, followed by evaluating the effects of genetic and chemical inhibition of Pin1 in leukemia cells on transformed phenotype, including cell proliferation and colony formation ability, using trypan blue, cell counting assay, and colony formation assay in vitro, as well as the tumorigenesis ability using in vivo xenograft mouse models. Results First, we found that the expression of Pin1 mRNA and protein was significantly increased in both de novo leukemia clinical samples and multiple leukemia cell lines, compared with healthy controls. Furthermore, genetic or chemical inhibition of Pin1 in human multiple leukemia cell lines potently inhibited multiple Pin1 substrate oncoproteins and effectively suppressed leukemia cell proliferation and colony formation ability in cell culture models in vitro. Moreover, tetracycline-inducible Pin1 knockdown and slow-releasing ATRA potently inhibited tumorigenicity of U937 and HL-60 leukemia cells in xenograft mouse models. Conclusions We demonstrate that Pin1 is highly overexpressed in human AML and is a promising therapeutic target to block multiple cancer-driving pathways in AML. Electronic supplementary material The online version of this article (10.1186/s13045-018-0611-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaolan Lian
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.,Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.,Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Yu-Min Lin
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Shingo Kozono
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Megan K Herbert
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Xin Li
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaohong Yuan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Jiangrui Guo
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Yafei Guo
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Min Tang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Jia Lin
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Yiping Huang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Bixin Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Chenxi Qiu
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Cheng-Yu Tsai
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jane Xie
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Ziang Jeff Gao
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Yong Wu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Hekun Liu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA. .,Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China.
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA. .,Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China.
| | - Yuanzhong Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
29
|
Matena A, Rehic E, Hönig D, Kamba B, Bayer P. Structure and function of the human parvulins Pin1 and Par14/17. Biol Chem 2018; 399:101-125. [PMID: 29040060 DOI: 10.1515/hsz-2017-0137] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022]
Abstract
Parvulins belong to the family of peptidyl-prolyl cis/trans isomerases (PPIases) assisting in protein folding and in regulating the function of a broad variety of proteins in all branches of life. The human representatives Pin1 and Par14/17 are directly involved in processes influencing cellular maintenance and cell fate decisions such as cell-cycle progression, metabolic pathways and ribosome biogenesis. This review on human parvulins summarizes the current knowledge of these enzymes and intends to oppose the well-studied Pin1 to its less well-examined homolog human Par14/17 with respect to structure, catalytic and cellular function.
Collapse
Affiliation(s)
- Anja Matena
- Structural and Medicinal Biochemistry, Center for Medical Biotechnology (ZMB), Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, D-45117 Essen, Germany
| | - Edisa Rehic
- Structural and Medicinal Biochemistry, Center for Medical Biotechnology (ZMB), Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, D-45117 Essen, Germany
| | - Dana Hönig
- Structural and Medicinal Biochemistry, Center for Medical Biotechnology (ZMB), Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, D-45117 Essen, Germany
| | - Bianca Kamba
- Structural and Medicinal Biochemistry, Center for Medical Biotechnology (ZMB), Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, D-45117 Essen, Germany
| | - Peter Bayer
- Structural and Medicinal Biochemistry, Center for Medical Biotechnology (ZMB), Faculty of Biology, University of Duisburg-Essen, Universitätsstr. 2, D-45117 Essen, Germany
| |
Collapse
|
30
|
Thomas AL, Lind H, Hong A, Dokic D, Oppat K, Rosenthal E, Guo A, Thomas A, Hamden R, Jeruss JS. Inhibition of CDK-mediated Smad3 phosphorylation reduces the Pin1-Smad3 interaction and aggressiveness of triple negative breast cancer cells. Cell Cycle 2017; 16:1453-1464. [PMID: 28678584 DOI: 10.1080/15384101.2017.1338988] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that lacks effective targeted therapies. Although TNBC is not defined by specific therapeutic targets, a subset of patients have tumors that overexpress cyclins. High cyclin D/E expression catalyzes CDK4/2 activity. In turn, CDK4/2 can non-canonically phosphorylate Smad3, a key TGFβ signaling intermediate, and this phosphorylation has been associated with the shift from tumor-suppressive to oncogenic TGFβ pathway action in breast oncogenesis. Additionally, CDK-mediated Smad3 phosphorylation facilitates an interaction between Smad3 and Pin1, a cis-trans isomerase that is also overexpressed in aggressive breast cancers. Treatment with CYC065, a CDK2/9 inhibitor, decreased non-canonical Smad3 phosphorylation and inhibited the Pin1-Smad3 interaction. We hypothesized that the interaction of Pin1 and Smad3, facilitated by CDK-mediated Smad3 phosphorylation, promotes TNBC cell aggressiveness. Inhibition of the Pin1-Smad3 interaction in TNBC cell lines, through depletion of Pin1 or CYC065 treatment, resulted in decreased cell migration/invasion and impeded the EMT program. Inhibition of CDK-mediated phosphorylation of Smad3 by mutagenesis also decreased cell migration, underscoring the importance of non-canonical CDK2 phosphorylation of Smad3 to enable cell motility. Pin1 depletion restored Smad3 protein levels and tumor-suppressive activity, suggesting that the Pin1-Smad3 interaction has a negative impact on canonical Smad3 action. Collectively, the data show that the Pin1-Smad3 interaction, facilitated by CDK-mediated Smad3 phosphorylation, is associated with oncogenic TGFβ signaling and breast cancer progression. Inhibition of this interaction with CYC065 treatment may provide an important therapeutic option for TNBC patients.
Collapse
Affiliation(s)
- Alexandra L Thomas
- a Driskill Graduate Program , Northwestern University , Chicago , IL , USA
| | - Hanne Lind
- b University of Michigan , Ann Arbor , MI , USA
| | - Angela Hong
- b University of Michigan , Ann Arbor , MI , USA
| | - Danijela Dokic
- c Department of Obstetrics and Gynecology , Northwestern University , Chicago , IL , USA
| | | | | | - Amina Guo
- b University of Michigan , Ann Arbor , MI , USA
| | - Aaron Thomas
- d Department of Surgery , University of Michigan , Ann Arbor , MI , USA
| | - Randala Hamden
- e Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Jacqueline S Jeruss
- d Department of Surgery , University of Michigan , Ann Arbor , MI , USA.,e Northwestern University Feinberg School of Medicine , Chicago , IL , USA.,f Department of Biomedical Engineering , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
31
|
Nakatsu Y, Mori K, Matsunaga Y, Yamamotoya T, Ueda K, Inoue Y, Mitsuzaki-Miyoshi K, Sakoda H, Fujishiro M, Yamaguchi S, Kushiyama A, Ono H, Ishihara H, Asano T. The prolyl isomerase Pin1 increases β-cell proliferation and enhances insulin secretion. J Biol Chem 2017; 292:11886-11895. [PMID: 28566287 DOI: 10.1074/jbc.m117.780726] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/23/2017] [Indexed: 11/06/2022] Open
Abstract
The prolyl isomerase Pin1 binds to the phosphorylated Ser/Thr-Pro motif of target proteins and enhances their cis-trans conversion. This report is the first to show that Pin1 expression in pancreatic β cells is markedly elevated by high-fat diet feeding and in ob/ob mice. To elucidate the role of Pin1 in pancreatic β cells, we generated β-cell-specific Pin1 KO (βPin1 KO) mice. These mutant mice showed exacerbation of glucose intolerance but had normal insulin sensitivity. We identified two independent factors underlying impaired insulin secretion in the βPin1 KO mice. Pin1 enhanced pancreatic β-cell proliferation, as indicated by a reduced β-cell mass in βPin1 KO mice compared with control mice. Moreover, a diet high in fat and sucrose failed to increase pancreatic β-cell growth in the βPin1 KO mice, an observation to which up-regulation of the cell cycle protein cyclin D appeared to contribute. The other role of Pin1 was to activate the insulin-secretory step: Pin1 KO β cells showed impairments in glucose- and KCl-induced elevation of the intracellular Ca2+ concentration and insulin secretion. We also identified salt-inducible kinase 2 (SIK2) as a Pin1-binding protein that affected the regulation of Ca2+ influx and found Pin1 to enhance SIK2 kinase activity, resulting in a decrease in p35 protein, a negative regulator of Ca2+ influx. Taken together, our observations demonstrate critical roles of Pin1 in pancreatic β cells and that Pin1 both promotes β-cell proliferation and activates insulin secretion.
Collapse
Affiliation(s)
- Yusuke Nakatsu
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Keiichi Mori
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Yasuka Matsunaga
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Takeshi Yamamotoya
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Koji Ueda
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Yuki Inoue
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Keiko Mitsuzaki-Miyoshi
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692
| | - Midori Fujishiro
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Itabashi, Tokyo 173-8610
| | - Suguru Yamaguchi
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Itabashi, Tokyo 173-8610
| | - Akifumi Kushiyama
- Division of Diabetes and Metabolism, Institute for Adult Diseases, Asahi Life Foundation, Chuo-ku, Tokyo 103-0002
| | - Hiraku Ono
- Department of Clinical Cell Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohara, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Hisamitsu Ishihara
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Itabashi, Tokyo 173-8610
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551.
| |
Collapse
|
32
|
Greseth MD, Carter DC, Terhune SS, Traktman P. Proteomic Screen for Cellular Targets of the Vaccinia Virus F10 Protein Kinase Reveals that Phosphorylation of mDia Regulates Stress Fiber Formation. Mol Cell Proteomics 2017; 16:S124-S143. [PMID: 28183815 PMCID: PMC5393388 DOI: 10.1074/mcp.m116.065003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/28/2017] [Indexed: 01/12/2023] Open
Abstract
Vaccinia virus, a complex dsDNA virus, is unusual in replicating exclusively within the cytoplasm of infected cells. Although this prototypic poxvirus encodes >200 proteins utilized during infection, a significant role for host proteins and cellular architecture is increasingly evident. The viral B1 kinase and H1 phosphatase are known to target cellular proteins as well as viral substrates, but little is known about the cellular substrates of the F10 kinase. F10 is essential for virion morphogenesis, beginning with the poorly understood process of diversion of membranes from the ER for the purpose of virion membrane biogenesis. To better understand the function of F10, we generated a cell line that carries a single, inducible F10 transgene. Using uninduced and induced cells, we performed stable isotope labeling of amino acids in cell culture (SILAC) coupled with phosphopeptide analysis to identify cellular targets of F10-mediated phosphorylation. We identified 27 proteins that showed statistically significant changes in phosphorylation upon the expression of the F10 kinase: 18 proteins showed an increase in phosphorylation whereas 9 proteins showed a decrease in phosphorylation. These proteins participate in several distinct cellular processes including cytoskeleton dynamics, membrane trafficking and cellular metabolism. One of the proteins with the greatest change in phosphorylation was mDia, a member of the formin family of cytoskeleton regulators; F10 induction led to increased phosphorylation on Ser22 Induction of F10 induced a statistically significant decrease in the percentage of cells with actin stress fibers; however, this change was abrogated when an mDia Ser22Ala variant was expressed. Moreover, expression of a Ser22Asp variant leads to a reduction of stress fibers even in cells not expressing F10. In sum, we present the first unbiased screen for cellular targets of F10-mediated phosphorylation, and in so doing describe a heretofore unknown mechanism for regulating stress fiber formation through phosphorylation of mDia. Data are available via ProteomeXchange with identifier PXD005246.
Collapse
Affiliation(s)
- Matthew D Greseth
- From the ‡Departments of Biochemistry & Molecular Biology and Microbiology & Immunology, and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Dominique C Carter
- §Department of Microbiology & Molecular Genetics and the Biotechnology & Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Scott S Terhune
- §Department of Microbiology & Molecular Genetics and the Biotechnology & Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Paula Traktman
- From the ‡Departments of Biochemistry & Molecular Biology and Microbiology & Immunology, and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina;
| |
Collapse
|
33
|
Liu C, Mu C, Li Z, Xu L. Imazamethabenz inhibits human breast cancer cell proliferation, migration and invasion via combination with Pin1. Mol Med Rep 2017; 15:3210-3214. [PMID: 28350086 DOI: 10.3892/mmr.2017.6399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
Abstract
Overexpression of peptidyl-prolyl cis/trans isomerase, NIMA interacting‑1 (Pin1) is a significant marker of the occurrence and development of tumors. In the present study, the imidazoline ketone herbicide imazamethabenz was investigated as a potential antitumor drug by inhibiting Pin1. Molecular docking and enzyme activity tests verified, for the first time, that the imidazoline ketone compound imazamethabenz effectively inhibited Pin1 in vitro. MTT assays, western blotting, wound healing assay and Matrigel invasion assays revealed that imazamethabenz induced apoptosis and inhibited migration and invasion of the breast cancer cell line MCF‑7, which overexpresses Pin1, by inhibiting the Pin1‑mediated signaling pathway involving vascular endothelial growth factor and matrix metalloproteinase 9. These findings indicated that imazamethabenz offers potential applications for the treatment of tumors as a Pin1 inhibitor.
Collapse
Affiliation(s)
- Chen Liu
- College of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Chaoyu Mu
- College of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zeng Li
- College of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Liang Xu
- College of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|