1
|
Ghislat G, Hernandez-Hernandez S, Piyawajanusorn C, Ballester PJ. Data-centric challenges with the application and adoption of artificial intelligence for drug discovery. Expert Opin Drug Discov 2024; 19:1297-1307. [PMID: 39316009 DOI: 10.1080/17460441.2024.2403639] [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: 07/09/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION Artificial intelligence (AI) is exhibiting tremendous potential to reduce the massive costs and long timescales of drug discovery. There are however important challenges currently limiting the impact and scope of AI models. AREAS COVERED In this perspective, the authors discuss a range of data issues (bias, inconsistency, skewness, irrelevance, small size, high dimensionality), how they challenge AI models, and which issue-specific mitigations have been effective. Next, they point out the challenges faced by uncertainty quantification techniques aimed at enhancing and trusting the predictions from these AI models. They also discuss how conceptual errors, unrealistic benchmarks and performance misestimation can confound the evaluation of models and thus their development. Lastly, the authors explain how human bias, whether from AI experts or drug discovery experts, constitutes another challenge that can be alleviated by gaining more prospective experience. EXPERT OPINION AI models are often developed to excel on retrospective benchmarks unlikely to anticipate their prospective performance. As a result, only a few of these models are ever reported to have prospective value (e.g. by discovering potent and innovative drug leads for a therapeutic target). The authors have discussed what can go wrong in practice with AI for drug discovery. The authors hope that this will help inform the decisions of editors, funders investors, and researchers working in this area.
Collapse
Affiliation(s)
- Ghita Ghislat
- Department of Life Sciences, Imperial College London, London, UK
| | | | | | | |
Collapse
|
2
|
Vishwakarma S, Hernandez-Hernandez S, Ballester PJ. Graph neural networks are promising for phenotypic virtual screening on cancer cell lines. Biol Methods Protoc 2024; 9:bpae065. [PMID: 39502795 PMCID: PMC11537795 DOI: 10.1093/biomethods/bpae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/20/2024] [Accepted: 09/02/2024] [Indexed: 11/08/2024] Open
Abstract
Artificial intelligence is increasingly driving early drug design, offering novel approaches to virtual screening. Phenotypic virtual screening (PVS) aims to predict how cancer cell lines respond to different compounds by focusing on observable characteristics rather than specific molecular targets. Some studies have suggested that deep learning may not be the best approach for PVS. However, these studies are limited by the small number of tested molecules as well as not employing suitable performance metrics and dissimilar-molecules splits better mimicking the challenging chemical diversity of real-world screening libraries. Here we prepared 60 datasets, each containing approximately 30 000-50 000 molecules tested for their growth inhibitory activities on one of the NCI-60 cancer cell lines. We conducted multiple performance evaluations of each of the five machine learning algorithms for PVS on these 60 problem instances. To provide even a more comprehensive evaluation, we used two model validation types: the random split and the dissimilar-molecules split. Overall, about 14 440 training runs aczross datasets were carried out per algorithm. The models were primarily evaluated using hit rate, a more suitable metric in VS contexts. The results show that all models are more challenged by test molecules that are substantially different from those in the training data. In both validation types, the D-MPNN algorithm, a graph-based deep neural network, was found to be the most suitable for building predictive models for this PVS problem.
Collapse
Affiliation(s)
- Sachin Vishwakarma
- Evotec SAS (France), Toulouse, France
- Centre de Recherche en Cancérologie de Marseille, Marseille 13009, France
| | | | - Pedro J Ballester
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
3
|
Bennett GM, Starczewski J, dela Cerna MVC. In silico identification of putative druggable pockets in PRL3, a significant oncology target. Biochem Biophys Rep 2024; 39:101767. [PMID: 39050014 PMCID: PMC11267023 DOI: 10.1016/j.bbrep.2024.101767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Protein tyrosine phosphatases (PTP) have emerged as targets in diseases characterized by aberrant phosphorylations such as cancers. The activity of the phosphatase of regenerating liver 3, PRL3, has been linked to several oncogenic and metastatic pathways, particularly in breast, ovarian, colorectal, and blood cancers. Development of small molecules that directly target PRL3, however, has been challenging. This is partly due to the lack of structural information on how PRL3 interacts with its inhibitors. Here, computational methods are used to bridge this gap by evaluating the druggability of PRL3. In particular, web-based pocket prediction tools, DoGSite3 and FTMap, were used to identify binding pockets using structures of PRL3 currently available in the Protein Data Bank. Druggability assessment by molecular dynamics simulations with probes was also performed to validate these results and to predict the strength of binding in the identified pockets. While several druggable pockets were identified, those in the closed conformation show more promise given their volume and depth. These two pockets flank the active site loops and roughly correspond to pockets predicted by molecular docking in previous papers. Notably, druggability simulations predict the possibility of low nanomolar affinity inhibitors in these sites implying the potential to identify highly potent small molecule inhibitors for PRL3. Putative pockets identified here can be leveraged for high-throughput virtual screening to further accelerate the drug discovery against PRL3 and development of PRL3-directed therapeutics.
Collapse
Affiliation(s)
- Grace M. Bennett
- Department of Biochemistry, Chemistry, and Physics, Georgia Southern University, Savannah, GA, 31419, USA
| | - Julia Starczewski
- Department of Biochemistry, Chemistry, and Physics, Georgia Southern University, Savannah, GA, 31419, USA
| | - Mark Vincent C. dela Cerna
- Department of Biochemistry, Chemistry, and Physics, Georgia Southern University, Savannah, GA, 31419, USA
| |
Collapse
|
4
|
Caba K, Tran-Nguyen VK, Rahman T, Ballester PJ. Comprehensive machine learning boosts structure-based virtual screening for PARP1 inhibitors. J Cheminform 2024; 16:40. [PMID: 38582911 PMCID: PMC10999096 DOI: 10.1186/s13321-024-00832-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/23/2024] [Indexed: 04/08/2024] Open
Abstract
Poly ADP-ribose polymerase 1 (PARP1) is an attractive therapeutic target for cancer treatment. Machine-learning scoring functions constitute a promising approach to discovering novel PARP1 inhibitors. Cutting-edge PARP1-specific machine-learning scoring functions were investigated using semi-synthetic training data from docking activity-labelled molecules: known PARP1 inhibitors, hard-to-discriminate decoys property-matched to them with generative graph neural networks and confirmed inactives. We further made test sets harder by including only molecules dissimilar to those in the training set. Comprehensive analysis of these datasets using five supervised learning algorithms, and protein-ligand fingerprints extracted from docking poses and ligand only features revealed one highly predictive scoring function. This is the PARP1-specific support vector machine-based regressor, when employing PLEC fingerprints, which achieved a high Normalized Enrichment Factor at the top 1% on the hardest test set (NEF1% = 0.588, median of 10 repetitions), and was more predictive than any other investigated scoring function, especially the classical scoring function employed as baseline.
Collapse
Affiliation(s)
- Klaudia Caba
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Viet-Khoa Tran-Nguyen
- Unité de Biologie Fonctionnelle et Adaptative (BFA), UFR Sciences du Vivant, Université Paris Cité, 75013, Paris, France
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Pedro J Ballester
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
5
|
Lin C, Yu M, Wu X, Wang H, Wei M, Zhang L. Targeting Moonlighting Enzymes in Cancer. Molecules 2024; 29:1573. [PMID: 38611852 PMCID: PMC11013064 DOI: 10.3390/molecules29071573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Moonlighting enzymes are multifunctional proteins that perform multiple functions beyond their primary role as catalytic enzymes. Extensive research and clinical practice have demonstrated their pivotal roles in the development and progression of cancer, making them promising targets for drug development. This article delves into multiple notable moonlighting enzymes, including GSK-3, GAPDH, and ENO1, and with a particular emphasis on an enigmatic phosphatase, PTP4A3. We scrutinize their distinct roles in cancer and the mechanisms that dictate their ability to switch roles. Lastly, we discuss the potential of an innovative approach to develop drugs targeting these moonlighting enzymes: target protein degradation. This strategy holds promise for effectively tackling moonlighting enzymes in the context of cancer therapy.
Collapse
Affiliation(s)
- Chunxu Lin
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Mingyang Yu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Ximei Wu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Hui Wang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Min Wei
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
6
|
Xiao S, Chen H, Bai Y, Zhang ZY, Liu Y. Targeting PRL phosphatases in hematological malignancies. Expert Opin Ther Targets 2024; 28:259-271. [PMID: 38653737 PMCID: PMC12050007 DOI: 10.1080/14728222.2024.2344695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION Phosphatase of regenerating liver (PRL) family proteins, also known as protein tyrosine phosphatase 4A (PTP4A), have been implicated in many types of cancers. The PRL family of phosphatases consists of three members, PRL1, PRL2, and PRL3. PRLs have been shown to harbor oncogenic potentials and are highly expressed in a variety of cancers. Given their roles in cancer progression and metastasis, PRLs are potential targets for anticancer therapies. However, additional studies are needed to be performed to fully understand the roles of PRLs in blood cancers. AREAS COVERED In this review, we will summarize recent studies of PRLs in normal and malignant hematopoiesis, the role of PRLs in regulating various signaling pathways, and the therapeutic potentials of targeting PRLs in hematological malignancies. We will also discuss how to improve current PRL inhibitors for cancer treatment. EXPERT OPINION Although PRL inhibitors show promising therapeutic effects in preclinical studies of different types of cancers, moving PRL inhibitors from bench to bedside is still challenging. More potent and selective PRL inhibitors are needed to target PRLs in hematological malignancies and improve treatment outcomes.
Collapse
Affiliation(s)
- Shiyu Xiao
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Hongxia Chen
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
- Department of Hematology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yunpeng Bai
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Institute for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, USA
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Institute for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, USA
| | - Yan Liu
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
7
|
Smith CN, Kihn K, Williamson ZA, Chow KM, Hersh LB, Korotkov KV, Deredge D, Blackburn JS. Development and characterization of nanobodies that specifically target the oncogenic Phosphatase of Regenerating Liver-3 (PRL-3) and impact its interaction with a known binding partner, CNNM3. PLoS One 2023; 18:e0285964. [PMID: 37220097 PMCID: PMC10204944 DOI: 10.1371/journal.pone.0285964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/04/2023] [Indexed: 05/25/2023] Open
Abstract
Phosphatase of Regenerating Liver-3 (PRL-3) is associated with cancer progression and metastasis. The mechanisms that drive PRL-3's oncogenic functions are not well understood, partly due to a lack of research tools available to study this protein. We have begun to address these issues by developing alpaca-derived single domain antibodies, or nanobodies, targeting PRL-3 with a KD of 30-300 nM and no activity towards highly homologous family members PRL-1 and PRL-2. We found that longer and charged N-terminal tags on PRL-3, such as GFP and FLAG, changed PRL-3 localization compared to untagged protein, indicating that the nanobodies may provide new insights into PRL-3 trafficking and function. The nanobodies perform equally, if not better, than commercially available antibodies in immunofluorescence and immunoprecipitation. Finally, hydrogen-deuterium exchange mass spectrometry (HDX-MS) showed that the nanobodies bind partially within the PRL-3 active site and can interfere with PRL-3 phosphatase activity. Co-immunoprecipitation with a known PRL-3 active site binding partner, the CBS domain of metal transporter CNNM3, showed that the nanobodies reduced the amount of PRL-3:CBS inter-action. The potential of blocking this interaction is highly relevant in cancer, as multiple research groups have shown that PRL-3 binding to CNNM proteins is sufficient to promote metastatic growth in mouse models. The anti-PRL-3 nanobodies represent an important expansion of the research tools available to study PRL-3 function and can be used to define the role of PRL-3 in cancer progression.
Collapse
Affiliation(s)
- Caroline N. Smith
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
- University of Kentucky Markey Cancer Center, Lexington, Kentucky, United States of America
| | - Kyle Kihn
- University of Maryland School of Pharmacy, Baltimore, Maryland, United States of America
| | - Zachary A. Williamson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - K. Martin Chow
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Louis B. Hersh
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Konstantin V. Korotkov
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Daniel Deredge
- University of Maryland School of Pharmacy, Baltimore, Maryland, United States of America
| | - Jessica S. Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
- University of Kentucky Markey Cancer Center, Lexington, Kentucky, United States of America
| |
Collapse
|
8
|
Chia PL, Ang KH, Thura M, Zeng Q. PRL3 as a therapeutic target for novel cancer immunotherapy in multiple cancer types. Theranostics 2023; 13:1876-1891. [PMID: 37064866 PMCID: PMC10091880 DOI: 10.7150/thno.79265] [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: 09/26/2022] [Accepted: 12/20/2022] [Indexed: 04/18/2023] Open
Abstract
Phosphatase of Regenerating Liver-3 (PRL3) was discovered in 1998 and was subsequently found to be correlated with cancer progression and metastasis in 2001. Extensive research in the past two decades has produced significant findings on PRL3-mediated cancer signaling and functions, as well as its clinical relevance in diverse types of cancer. PRL3 has been established to play a role in many cancer-related functions, including but not limited to metastasis, proliferation, and angiogenesis. Importantly, the tumor-specific expression of PRL3 protein in multiple cancer types has made it an attractive therapeutic target. Much effort has been made in developing PRL3-targeted therapy with small chemical inhibitors against intracellular PRL3, and notably, the development of PRL3-zumab as a novel cancer immunotherapy against PRL3. In this review, we summarize the current understanding of the role of PRL3 in cancer-related cellular functions, its prognostic value, as well as perspectives on PRL3 as a target for unconventional immunotherapy in the clinic with PRL3-zumab.
Collapse
Affiliation(s)
- Pei Ling Chia
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore 138673; ; ;
| | - Koon Hwee Ang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore 138673; ; ;
| | - Min Thura
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore 138673; ; ;
| | - Qi Zeng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore 138673; ; ;
| |
Collapse
|
9
|
The phosphatase PRL-3 affects intestinal homeostasis by altering the crypt cell composition. J Mol Med (Berl) 2021; 99:1413-1426. [PMID: 34129057 PMCID: PMC8455404 DOI: 10.1007/s00109-021-02097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/04/2021] [Accepted: 05/20/2021] [Indexed: 11/04/2022]
Abstract
Expression of the phosphatase of regenerating liver-3 (PRL-3) is known to promote tumor growth in gastrointestinal adenocarcinomas, and the incidence of tumor formation upon inflammatory events correlates with PRL-3 levels in mouse models. These carcinomas and their onset are associated with the impairment of intestinal cell homeostasis, which is regulated by a balanced number of Paneth cells and Lgr5 expressing intestinal stem cells (Lgr5+ ISCs). Nevertheless, the consequences of PRL-3 overexpression on cellular homeostasis and ISC fitness in vivo are unexplored. Here, we employ a doxycycline-inducible PRL-3 mouse strain to show that aberrant PRL-3 expression within a non-cancerous background leads to the death of Lgr5+ ISCs and to Paneth cell expansion. A higher dose of PRL-3, resulting from homozygous expression, led to mice dying early. A primary 3D intestinal culture model obtained from these mice confirmed the loss of Lgr5+ ISCs upon PRL-3 expression. The impaired intestinal organoid formation was rescued by a PRL inhibitor, providing a functional link to the observed phenotypes. These results demonstrate that elevated PRL-3 phosphatase activity in healthy intestinal epithelium impairs intestinal cell homeostasis, which correlates this cellular mechanism of tumor onset with PRL-3-mediated higher susceptibility to tumor formation upon inflammatory or mutational events. Key messages • Transgenic mice homozygous for PRL-3 overexpression die early. • PRL-3 heterozygous mice display disrupted intestinal self-renewal capacity. • PRL-3 overexpression alone does not induce tumorigenesis in the mouse intestine. • PRL-3 activity leads to the death of Lgr5+ ISCs and Paneth cell expansion. • Impairment of cell homeostasis correlates PRL-3 action with tumor onset mechanisms.
Collapse
|
10
|
A screen of FDA-approved drugs identifies inhibitors of protein tyrosine phosphatase 4A3 (PTP4A3 or PRL-3). Sci Rep 2021; 11:10302. [PMID: 33986418 PMCID: PMC8119466 DOI: 10.1038/s41598-021-89668-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 04/29/2021] [Indexed: 12/02/2022] Open
Abstract
Protein tyrosine phosphatase 4A3 (PTP4A3 or PRL-3) is highly expressed in a variety of cancers, where it promotes tumor cell migration and metastasis leading to poor prognosis. Despite its clinical significance, small molecule inhibitors of PRL-3 are lacking. Here, we screened 1443 FDA-approved drugs for their ability to inhibit the activity of the PRL phosphatase family. We identified five specific inhibitors for PRL-3 as well as one selective inhibitor of PRL-2. Additionally, we found nine drugs that broadly and significantly suppressed PRL activity. Two of these broad-spectrum PRL inhibitors, Salirasib and Candesartan, blocked PRL-3-induced migration in human embryonic kidney cells with no impact on cell viability. Both drugs prevented migration of human colorectal cancer cells in a PRL-3 dependent manner and were selective towards PRLs over other phosphatases. In silico modeling revealed that Salirasib binds a putative allosteric site near the WPD loop of PRL-3, while Candesartan binds a potentially novel targetable site adjacent to the CX5R motif. Inhibitor binding at either of these sites is predicted to trap PRL-3 in a closed conformation, preventing substrate binding and inhibiting function.
Collapse
|
11
|
|
12
|
Cai F, Huang Y, Wang M, Sun M, Zhao Y, Hattori M. A FRET-based screening method to detect potential inhibitors of the binding of CNNM3 to PRL2. Sci Rep 2020; 10:12879. [PMID: 32733084 PMCID: PMC7393355 DOI: 10.1038/s41598-020-69818-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/08/2020] [Indexed: 11/09/2022] Open
Abstract
The cyclin M (CNNM) family of Mg2+ transporters is reported to promote tumour progression by binding to phosphatase of regenerating liver (PRL) proteins. Here, we established an assay for detection of the binding between the cystathionine-beta-synthase (CBS) domain of human CNNM3 (a region responsible for PRL binding) and human PRL2 using fluorescence resonance energy transfer (FRET) techniques. By fusing YPet to the C-terminus of the CNNM3 CBS domain and CyPet to the N-terminus of PRL2, we performed a FRET-based binding assay with purified proteins in multiwell plates and successfully detected the changes in fluorescence intensity derived from FRET with a reasonable Kd. We then confirmed that the addition of non-YPet-tagged CNNM3 and non-CyPet-tagged PRL proteins inhibited the changes in FRET intensity, whereas non-YPet-tagged CNNM3 with a mutation at the PRL2-binding site did not exhibit such inhibition. Furthermore, newly synthesized peptides derived from the CNNM loop region, with the PRL-binding sequences of the CNNM3 CBS domain, inhibited the interactions between CNNM3 and PRL2. Overall, these results showed that this method can be used for screening to identify inhibitors of CNNM-PRL interactions, potentially for novel anticancer therapy.
Collapse
Affiliation(s)
- Faji Cai
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200438, China
| | - Yichen Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200438, China
| | - Mengqi Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200438, China
| | - Minxuan Sun
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200438, China
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200438, China.
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200438, China.
| |
Collapse
|
13
|
Köhn M. Turn and Face the Strange: A New View on Phosphatases. ACS CENTRAL SCIENCE 2020; 6:467-477. [PMID: 32341996 PMCID: PMC7181316 DOI: 10.1021/acscentsci.9b00909] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Indexed: 05/08/2023]
Abstract
Phosphorylation as a post-translational modification is critical for cellular homeostasis. Kinases and phosphatases regulate phosphorylation levels by adding or removing, respectively, a phosphate group from proteins or other biomolecules. Imbalances in phosphorylation levels are involved in a multitude of diseases. Phosphatases are often thought of as the black sheep, the strangers, of phosphorylation-mediated signal transduction, particularly when it comes to drug discovery and development. This is due to past difficulties to study them and unsuccessful attempts to target them; however, phosphatases have regained strong attention and are actively pursued now in clinical trials. By giving examples for current hot topics in phosphatase biology and for new approaches to target them, it is illustrated here how and why phosphatases made their comeback, and what is envisioned to come in the future.
Collapse
Affiliation(s)
- Maja Köhn
- Faculty
of Biology, Institute of Biology III, University
of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Signalling
Research Centres BIOSS and CIBSS, University
of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Wei M, Haney MG, Rivas DR, Blackburn JS. Protein tyrosine phosphatase 4A3 (PTP4A3/PRL-3) drives migration and progression of T-cell acute lymphoblastic leukemia in vitro and in vivo. Oncogenesis 2020; 9:6. [PMID: 32001668 PMCID: PMC6992623 DOI: 10.1038/s41389-020-0192-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/23/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive blood cancer. There are no immunotherapies and few molecularly targeted therapeutics available for treatment of this malignancy. The identification and characterization of genes and pathways that drive T-ALL progression are critical for the development of new therapies for T-ALL. Here, we determined that the protein tyrosine phosphatase 4A3 (PTP4A3 or PRL-3) plays a critical role in T-ALL initiation and progression by promoting leukemia cell migration. PRL-3 is highly expressed in patient T-ALL samples at both the mRNA and protein levels compared to normal lymphocytes. Knock-down of PRL-3 expression using short-hairpin RNA (shRNA) in human T-ALL cell lines significantly impeded T-ALL cell migration capacity in vitro and reduced their ability to engraft and proliferate in vivo in xenograft mouse models. Additionally, PRL-3 overexpression in a Myc-induced zebrafish T-ALL model significantly accelerated disease onset and shortened the time needed for cells to enter blood circulation. Reverse-phase protein array (RPPA) and gene set enrichment analysis (GSEA) revealed that the SRC signaling pathway is affected by PRL-3. Immunoblot analyses validated that manipulation of PRL-3 expression in T-ALL cells affected the SRC signaling pathway, which is directly involved in cell migration, although Src was not a direct substrate of PRL-3. More importantly, T-ALL cell growth and migration were inhibited by small molecule inhibition of PRL-3, suggesting that PRL-3 has potential as a therapeutic target in T-ALL. Taken together, our study identifies PRL-3 as an oncogenic driver in T-ALL both in vitro and in vivo and provides a strong rationale for targeted therapies that interfere with PRL-3 function.
Collapse
Affiliation(s)
- M Wei
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 4053, USA
| | - M G Haney
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 4053, USA
- Markey Cancer Center, Lexington, KY, 40536, USA
| | - D R Rivas
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 4053, USA
| | - J S Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 4053, USA.
- Markey Cancer Center, Lexington, KY, 40536, USA.
| |
Collapse
|
15
|
Kharas GB, Cimino A, Flieger S, Whelpley PM, Ebner D, Groy R, Savittieri CR, Shinde N, Thomas KL, Rocus SM, Schjerven WS. Synthesis and vinyl benzene copolymerization of novel trisubstituted ethylenes: 15. Halogen and methoxy ring-substituted isopropyl 2-cyano-3-phenyl-2-propenoates. Des Monomers Polym 2020; 23:75-82. [PMID: 33029076 PMCID: PMC7473155 DOI: 10.1080/15685551.2020.1782556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Condensation of isopropyl cyanoacetate and substituted benzoic aldehydes resulted in formation of novel isopropyl esters of 2-cyano-3-phenyl-2-propenoic acid, RPhCH = C(CN)CO2CH(CH3)2 (where R is 2,3,4-trimethoxy, 2,4,5-trimethoxy, 2,4,6-trimethoxy, 3-bromo-4,5-dimethoxy, 5-bromo-2,3-dimethoxy, 5-bromo-2,4-dimethoxy, 6-bromo-3,4-dimethoxy, 2-bromo-3-hydroxy-4-methoxy, 4-bromo-2,6-difluoro, 2-chloro-3,4-dimethoxy, 3-chloro-4,5-dimethoxy, 5-chloro-2,3-dimethoxy, 2,3,6-trichloro, 3-chloro-2,6-difluoro, 2,3,4-trifluoro, 2,4,5-trifluoro, 2,4,6-trifluoro, 3,4,5-trifluoro, 2,3,5,6-tetrafluoro, 2,3,4,5,6-pentafluoro). Copolymerization of the esters with vinyl benzene in solution with radical initiation (ABCN) at 70°C led to formation copolymers. The products were characterized by CHN elemental analysis, IR, 1 H- and 13 C-NMR, GPC, DSC, and TGA.
Collapse
Affiliation(s)
- Gregory B. Kharas
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - Alessandra Cimino
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - Sebastian Flieger
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - Paige M. Whelpley
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - David Ebner
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - Randi Groy
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | | | - Nita Shinde
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - Kenneth L. Thomas
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | - Sara M. Rocus
- Chemistry and Biochemistry Department, DePaul University, Chicago, IL, USA
| | | |
Collapse
|
16
|
Lazo JS, Blanco IK, Tasker NR, Rastelli EJ, Burnett JC, Garrott SR, Hart DJ, McCloud RL, Hsu KL, Wipf P, Sharlow ER. Next-Generation Cell-Active Inhibitors of the Undrugged Oncogenic PTP4A3 Phosphatase. J Pharmacol Exp Ther 2019; 371:652-662. [PMID: 31601683 PMCID: PMC6856870 DOI: 10.1124/jpet.119.262188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022] Open
Abstract
Oncogenic protein tyrosine phosphatases (PTPs) are overexpressed in numerous human cancers but they have been challenging pharmacological targets. The emblematic oncogenic PTP4A tyrosine phosphatase family regulates many fundamental malignant processes. 7-Imino-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione (JMS-053) is a novel, potent, and selective PTP4A inhibitor but its mechanism of action has not been fully elucidated, nor has the chemotype been fully investigated. Because tyrosine phosphatases are notoriously susceptible to oxidation, we interrogated JMS-053 and three newly synthesized analogs with specific attention on the role of oxidation. JMS-053 and its three analogs were potent in vitro PTP4A3 inhibitors, but 7-imino-5-methyl-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione (NRT-870-59) appeared unique among the thienopyridinediones with respect to its inhibitory specificity for PTP4A3 versus both a PTP4A3 A111S mutant and an oncogenic dual specificity tyrosine phosphatase, CDC25B. Like JMS-053, NRT-870-59 was a reversible PTP4A3 inhibitor. All of the thienopyridinediones retained cytotoxicity against human ovarian and breast cancer cells grown as pathologically relevant three-dimensional spheroids. Inhibition of cancer cell colony formation by NRT-870-59, like JMS-053, required PTP4A3 expression. JMS-053 failed to generate significant detectable reactive oxygen species in vitro or in cancer cells. Mass spectrometry results indicated no disulfide bond formation or oxidation of the catalytic Cys104 after in vitro incubation of PTP4A3 with JMS-053 or NRT-870-59. Gene expression profiling of cancer cells exposed to JMS-053 phenocopied many of the changes seen with the loss of PTP4A3 and did not indicate oxidative stress. These data demonstrate that PTP4A phosphatases can be selectively targeted with small molecules that lack prominent reactive oxygen species generation and encourage further studies of this chemotype. SIGNIFICANCE STATEMENT: Protein tyrosine phosphatases are emerging as important contributors to human cancers. We report on a new class of reversible protein phosphatase small molecule inhibitors that are cytotoxic to human ovarian and breast cancer cells, do not generate significant reactive oxygen species in vitro and in cells, and could be valuable lead molecules for future studies of PTP4A phosphatases.
Collapse
Affiliation(s)
- John S Lazo
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Isabella K Blanco
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nikhil R Tasker
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ettore J Rastelli
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James C Burnett
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sharon R Garrott
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Duncan J Hart
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rebecca L McCloud
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ku-Lung Hsu
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Peter Wipf
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elizabeth R Sharlow
- Departments of Pharmacology (J.S.L., I.K.B., S.R.G., D.J.H., E.R.S.) and Chemistry (J.S.L., R.L.M., K.-L.H.), University of Virginia, Charlottesville, Virginia; and Department of Chemistry (N.R.T., E.J.R., J.C.B., P.W.), University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
Ruckert MT, de Andrade PV, Santos VS, Silveira VS. Protein tyrosine phosphatases: promising targets in pancreatic ductal adenocarcinoma. Cell Mol Life Sci 2019; 76:2571-2592. [PMID: 30982078 PMCID: PMC11105579 DOI: 10.1007/s00018-019-03095-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer. It is the fourth leading cause of cancer-related death and is associated with a very poor prognosis. KRAS driver mutations occur in approximately 95% of PDAC cases and cause the activation of several signaling pathways such as mitogen-activated protein kinase (MAPK) pathways. Regulation of these signaling pathways is orchestrated by feedback loops mediated by the balance between protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs), leading to activation or inhibition of its downstream targets. The human PTPome comprises 125 members, and these proteins are classified into three distinct families according to their structure. Since PTP activity description, it has become clear that they have both inhibitory and stimulatory effects on cancer-associated signaling processes and that deregulation of PTP function is closely associated with tumorigenesis. Several PTPs have displayed either tumor suppressor or oncogenic characteristics during the development and progression of PDAC. In this sense, PTPs have been presented as promising candidates for the treatment of human pancreatic cancer, and many PTP inhibitors have been developed since these proteins were first associated with cancer. Nevertheless, some challenges persist regarding the development of effective and safe methods to target these molecules and deliver these drugs. In this review, we discuss the role of PTPs in tumorigenesis as tumor suppressor and oncogenic proteins. We have focused on the differential expression of these proteins in PDAC, as well as their clinical implications and possible targeting for pharmacological inhibition in cancer therapy.
Collapse
Affiliation(s)
- Mariana Tannús Ruckert
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Pamela Viani de Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Verena Silva Santos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Vanessa Silva Silveira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
18
|
Tasker NR, Rastelli EJ, Burnett JC, Sharlow ER, Lazo JS, Wipf P. Tapping the therapeutic potential of protein tyrosine phosphatase 4A with small molecule inhibitors. Bioorg Med Chem Lett 2019; 29:2008-2015. [PMID: 31307888 DOI: 10.1016/j.bmcl.2019.06.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022]
Abstract
Protein tyrosine phosphatases (PTPs) are emerging new targets for drug discovery. PTPs and protein tyrosine kinases (PTKs) maintain cellular homeostasis through opposing roles: tyrosine O-dephosphorylation and -phosphorylation, respectively. An imbalance in the phosphorylation equilibrium results in aberrant protein signaling and pathophysiological conditions. PTPs have historically been considered 'undruggable', in part due to a lack of evidence defining their relationship to disease causality and a focus on purely competitive inhibitors. However, a better understanding of protein-protein interfaces and shallow active sites has recently renewed interest in the pursuit of allosteric and orthosteric modulators of targets outside the major druggable protein families. While their biological mechanism of action still remains to be clarified, PTP4A1-3 (also referred to as PRL1-3) are validated oncology targets and play an important role in cell proliferation, metastasis, and tumor angiogenesis. In this Digest, recent syntheses and structure-activity relationships (SAR) of small molecule inhibitors (SMIs) of PTP4A1-3 are summarized, and enzyme docking studies of the most potent chemotype are highlighted. In particular, the thienopyridone scaffold has emerged as a potent lead structure to interrogate the function and druggability of this dual-specificity PTP.
Collapse
Affiliation(s)
- Nikhil R Tasker
- University of Pittsburgh, Department of Chemistry, 219 Parkman Avenue, Pittsburgh, PA 15260, USA
| | - Ettore J Rastelli
- University of Pittsburgh, Department of Chemistry, 219 Parkman Avenue, Pittsburgh, PA 15260, USA
| | - James C Burnett
- University of Pittsburgh, Department of Chemistry, 219 Parkman Avenue, Pittsburgh, PA 15260, USA
| | - Elizabeth R Sharlow
- University of Virginia, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - John S Lazo
- University of Virginia, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Peter Wipf
- University of Pittsburgh, Department of Chemistry, 219 Parkman Avenue, Pittsburgh, PA 15260, USA.
| |
Collapse
|
19
|
Peón A, Li H, Ghislat G, Leung KS, Wong MH, Lu G, Ballester PJ. MolTarPred: A web tool for comprehensive target prediction with reliability estimation. Chem Biol Drug Des 2019; 94:1390-1401. [PMID: 30916462 DOI: 10.1111/cbdd.13516] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/07/2019] [Accepted: 03/03/2019] [Indexed: 12/17/2022]
Abstract
Molecular target prediction can provide a starting point to understand the efficacy and side effects of phenotypic screening hits. Unfortunately, the vast majority of in silico target prediction methods are not available as web tools. Furthermore, these are limited in the number of targets that can be predicted, do not estimate which target predictions are more reliable and/or lack comprehensive retrospective validations. We present MolTarPred ( http://moltarpred.marseille.inserm.fr/), a user-friendly web tool for predicting protein targets of small organic compounds. It is powered by a large knowledge base comprising 607,659 compounds and 4,553 macromolecular targets collected from the ChEMBL database. In about 1 min, the predicted targets for the supplied molecule will be listed in a table. The chemical structures of the query molecule and the most similar compounds annotated with the predicted target will also be shown to permit visual inspection and comparison. Practical examples of the use of MolTarPred are showcased. MolTarPred is a new resource for scientists that require a more complete knowledge of the polypharmacology of a molecule. The introduction of a reliability score constitutes an attractive functionality of MolTarPred, as it permits focusing experimental confirmatory tests on the most reliable predictions, which leads to higher prospective hit rates.
Collapse
Affiliation(s)
- Antonio Peón
- Centre de Recherche en Cancérologie de Marseille (CRCM), U1068, Inserm, Marseille, France.,UMR7258, CNRS, Marseille, France.,Institut Paoli-Calmettes, Marseille, France.,UM 105, Aix-Marseille University, Marseille, France
| | - Hongjian Li
- SDIVF R&D Centre, Hong Kong Science Park, Sha Tin, New Territories, Hong Kong.,CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong
| | - Ghita Ghislat
- U1104, CNRS UMR7280, Centre d'Immunologie de Marseille-Luminy, Inserm, Marseille, France
| | - Kwong-Sak Leung
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong
| | - Man-Hon Wong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong
| | - Pedro J Ballester
- Centre de Recherche en Cancérologie de Marseille (CRCM), U1068, Inserm, Marseille, France.,UMR7258, CNRS, Marseille, France.,Institut Paoli-Calmettes, Marseille, France.,UM 105, Aix-Marseille University, Marseille, France
| |
Collapse
|
20
|
Zhang Z, Kozlov G, Chen YS, Gehring K. Mechanism of thienopyridone and iminothienopyridinedione inhibition of protein phosphatases. MEDCHEMCOMM 2019; 10:791-799. [PMID: 31191869 DOI: 10.1039/c9md00175a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Abstract
Thienopyridone (TP) has been proposed as a selective inhibitor of phosphatases of regenerating liver (PRL or PTP4A). PRLs are dual specificity phosphatases that promote cancer progression and are attractive anticancer targets. TP and iminothienopyridinedione (ITP), a more potent derivative, were shown to be effective inhibitors but the mechanism of inhibition was not established. Here, we perform NMR experiments and in vitro phosphatase assays to show that TP and ITP inhibit protein phosphatases non-specifically through oxidation of the phosphatase catalytic cysteine. We demonstrate that TP and ITP are redox active compounds, inhibiting PRL-3 and multiple other PTPs through oxidation. They also catalyze the oxidation of thioredoxin-1 as well as small molecules, like TCEP, DTT, and glutathione. The reported selectivity of TP and ITP is likely due to the higher susceptibility of PRLs to oxidation. Thus, while TP and ITP effectively inhibit PRLs, their use for studying the cellular function of PRLs is problematic due to the likelihood of off-target effects.
Collapse
Affiliation(s)
- Zhidian Zhang
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| | - Guennadi Kozlov
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| | - Yu Seby Chen
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| | - Kalle Gehring
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| |
Collapse
|
21
|
Castro-Sánchez P, Ramirez-Munoz R, Martín-Cófreces NB, Aguilar-Sopeña O, Alegre-Gomez S, Hernández-Pérez S, Reyes R, Zeng Q, Cabañas C, Sánchez-Madrid F, Roda-Navarro P. Phosphatase of Regenerating Liver-1 (PRL-1) Regulates Actin Dynamics During Immunological Synapse Assembly and T Cell Effector Function. Front Immunol 2018; 9:2655. [PMID: 30515156 PMCID: PMC6255827 DOI: 10.3389/fimmu.2018.02655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022] Open
Abstract
The regulatory role of most dual specific phosphatases during T cell activation remains unknown. Here, we have studied the expression and function of phosphatases of regenerating liver (PRLs: PRL-1, PRL-2, and PRL-3) during T cell activation, as well as, the dynamic delivery of PRL-1 to the Immunological Synapse (IS). We found that T cell activation downregulates the expression of PRL-2, resulting in an increased PRL-1/PRL-2 ratio. PRL-1 redistributed at the IS in two stages: Initially, it was transiently accumulated at scanning membranes enriched in CD3 and actin, and at later times, it was delivered at the contact site from pericentriolar, CD3ζ-containing, vesicles. Once at the established IS, PRL-1 distributed to LFA-1 and CD3ε sites. Remarkably, PRL-1 was found to regulate actin dynamics during IS assembly and the secretion of IL-2. Moreover, pharmacological inhibition of the catalytic activity of the three PRLs reduced the secretion of IL-2. These results provide evidence indicating a regulatory role of PRL-1 during IS assembly and highlight the involvement of PRLs in immune responses by mature T cells.
Collapse
Affiliation(s)
- Patricia Castro-Sánchez
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Rocío Ramirez-Munoz
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Noa B Martín-Cófreces
- Servicio de Inmunología. Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Oscar Aguilar-Sopeña
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Sergio Alegre-Gomez
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Sara Hernández-Pérez
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Raquel Reyes
- Department of Cell Biology and Immunology, Center for Molecular Biology Severo Ochoa (CBM-SO), Mayor Council of Scientific Research (CSIC), Madrid, Spain
| | - Qi Zeng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Carlos Cabañas
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain.,Department of Cell Biology and Immunology, Center for Molecular Biology Severo Ochoa (CBM-SO), Mayor Council of Scientific Research (CSIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología. Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| |
Collapse
|
22
|
Kumar A, Zhang KYJ. Shape similarity guided pose prediction: lessons from D3R Grand Challenge 3. J Comput Aided Mol Des 2018; 33:47-59. [PMID: 30084081 DOI: 10.1007/s10822-018-0142-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022]
Abstract
To extend the utility of ligand 3D shape similarity into pose prediction and virtual screening, we have previously developed CDVS and PoPSS methods. Both of them utilize ligand 3D shape similarity with the crystallographic ligands to improve pose prediction. While CDVS utilizes shape similarity to select suitable receptor structures for molecular docking, PoPSS places a ligand conformation of the highest shape similarity with crystal ligands into the target protein binding pocket which is then refined by side-chain repacking and Monte Carlo energy minimization. Analyses of PoPSS revealed some drawbacks in ligand conformation generation and the scoring scheme used. Moreover, as PoPSS does not sample the ligand conformation after placing it in the binding pocket, it relies solely on conformation generation methods to produce native like conformations. To address these limitations of PoPSS method, we report here a modified approach named as PoPSS-Lite, where side-chain repacking was replaced by a simple grid-based energy minimization. This modification also allowed the sampling of terminal functional groups while keeping the core scaffold fixed. Furthermore, shape similarity calculations were improved by increasing the number of ligand conformations and using a different similarity metric. The performance of PoPSS-Lite was prospectively evaluated in D3R GC3. Comparison of PoPSS-Lite demonstrated superior performance over PoPSS and CDVS with lower mean and median RMSDs. Furthermore, comparison with other D3R GC3 pose prediction submissions revealed top performance for PoPSS-Lite. Our D3R GC3 result extends our perspective that ligand 3D shape similarity with known crystallographic information can be successfully used to predict the binding pose of ligands with unknown binding modes. Our D3R GC3 results further highlight the necessity for improvement in conformer generation methods in order to improve shape similarity guided pose prediction.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
23
|
Kumar A, Zhang KYJ. Advances in the Development of Shape Similarity Methods and Their Application in Drug Discovery. Front Chem 2018; 6:315. [PMID: 30090808 PMCID: PMC6068280 DOI: 10.3389/fchem.2018.00315] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022] Open
Abstract
Molecular similarity is a key concept in drug discovery. It is based on the assumption that structurally similar molecules frequently have similar properties. Assessment of similarity between small molecules has been highly effective in the discovery and development of various drugs. Especially, two-dimensional (2D) similarity approaches have been quite popular due to their simplicity, accuracy and efficiency. Recently, the focus has been shifted toward the development of methods involving the representation and comparison of three-dimensional (3D) conformation of small molecules. Among the 3D similarity methods, evaluation of shape similarity is now gaining attention for its application not only in virtual screening but also in molecular target prediction, drug repurposing and scaffold hopping. A wide range of methods have been developed to describe molecular shape and to determine the shape similarity between small molecules. The most widely used methods include atom distance-based methods, surface-based approaches such as spherical harmonics and 3D Zernike descriptors, atom-centered Gaussian overlay based representations. Several of these methods demonstrated excellent virtual screening performance not only retrospectively but also prospectively. In addition to methods assessing the similarity between small molecules, shape similarity approaches have been developed to compare shapes of protein structures and binding pockets. Additionally, shape comparisons between atomic models and 3D density maps allowed the fitting of atomic models into cryo-electron microscopy maps. This review aims to summarize the methodological advances in shape similarity assessment highlighting advantages, disadvantages and their application in drug discovery.
Collapse
Affiliation(s)
| | - Kam Y. J. Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Yokohama, Japan
| |
Collapse
|
24
|
Abstract
The phosphatase of regenerating liver (PRL) family, also known as protein tyrosine phosphatase 4A (PTP4A), are dual-specificity phosphatases with largely unknown cellular functions. However, accumulating evidence indicates that PRLs are oncogenic across a broad variety of human cancers. PRLs are highly expressed in advanced tumors and metastases compared to early stage cancers or matched healthy tissue, and high expression of PRLs often correlates with poor patient prognosis. Consequentially, PRLs have been considered potential therapeutic targets in cancer. Persistent efforts have been made to define their role and mechanism in cancer progression and to create specific PRL inhibitors for basic research and drug development. However, targeting PRLs with small molecules remains challenging due to the highly conserved active site of protein tyrosine phosphatases and a high degree of sequence similarity between the PRL protein families. Here, we review the current PRL inhibitors, including the strategies used for their identification, their biological efficacy, potency, and selectivity, with a special focus on how PRL structure can inform future efforts to develop specific PRL inhibitors.
Collapse
Affiliation(s)
- Min Wei
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Konstantin V Korotkov
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Jessica S Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
25
|
McQueeney KE, Salamoun JM, Ahn JG, Pekic P, Blanco IK, Struckman HL, Sharlow ER, Wipf P, Lazo JS. A chemical genetics approach identifies PTP4A3 as a regulator of colon cancer cell adhesion. FASEB J 2018; 32:5661-5673. [PMID: 29746167 DOI: 10.1096/fj.201701446r] [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] [Indexed: 12/19/2022]
Abstract
Dysregulation of the tightly controlled protein phosphorylation networks that govern cellular behavior causes cancer. The membrane-associated, intracellular protein tyrosine phosphatase PTP4A3 is overexpressed in human colorectal cancer and contributes to cell migration and invasion. To interrogate further the role of PTP4A3 in colorectal cancer cell migration and invasion, we deleted the Ptp4a3 gene from murine colorectal tumor cells. The resulting PTP4A3-/- cells exhibited impaired colony formation, spheroid formation, migration, and adherence compared with the paired PTP4A3fl/fl cells. We replicated these phenotypic changes using the new small-molecule, allosteric PTP4A3 inhibitor JMS-053. A related structure, JMS-038, which lacked phosphatase inhibition, displayed no cellular activity. Reduction in cell viability and colony formation by JMS-053 occurred in both mouse and human colorectal cell lines and required PTP4A3 expression. Ptp4a3 deletion increased the expression of extracellular matrix (ECM) and adhesion genes, including the tumor suppressor Emilin 1. JMS-053 also increased Emilin 1 gene expression. Moreover, The Cancer Genome Atlas genomic database revealed human colorectal tumors with high Ptp4a3 expression had low Emilin 1 expression. These chemical and biologic reagents reveal a previously unknown communication between the intracellular PTP4A3 phosphatase and the ECM and support efforts to pharmacologically target PTP4A3.-McQueeney, K. E., Salamoun, J. M., Ahn J. G., Pekic, P., Blanco, I. K., Struckman, H. L., Sharlow, E. R., Wipf, P., Lazo, J. S. A chemical genetics approach identifies PTP4A3 as a regulator of colon cancer cell adhesion.
Collapse
Affiliation(s)
- Kelley E McQueeney
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Joseph M Salamoun
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jennifer G Ahn
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Paula Pekic
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Isabella K Blanco
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Heather L Struckman
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth R Sharlow
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John S Lazo
- Fiske Drug Discovery Laboratory, Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
26
|
Buiga P, Elson A, Tabernero L, Schwartz JM. Regulation of dual specificity phosphatases in breast cancer during initial treatment with Herceptin: a Boolean model analysis. BMC SYSTEMS BIOLOGY 2018; 12:11. [PMID: 29671404 PMCID: PMC5907139 DOI: 10.1186/s12918-018-0534-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background 25% of breast cancer patients suffer from aggressive HER2-positive tumours that are characterised by overexpression of the HER2 protein or by its increased tyrosine kinase activity. Herceptin is a major drug used to treat HER2 positive breast cancer. Understanding the molecular events that occur when breast cancer cells are exposed to Herceptin is therefore of significant importance. Dual specificity phosphatases (DUSPs) are central regulators of cell signalling that function downstream of HER2, but their role in the cellular response to Herceptin is mostly unknown. This study aims to model the initial effects of Herceptin exposure on DUSPs in HER2-positive breast cancer cells using Boolean modelling. Results We experimentally measured expression time courses of 21 different DUSPs between 0 and 24 h following Herceptin treatment of human MDA-MB-453 HER2-positive breast cancer cells. We clustered these time courses into patterns of similar dynamics over time. In parallel, we built a series of Boolean models representing the known regulatory mechanisms of DUSPs and then demonstrated that the dynamics predicted by the models is in agreement with the experimental data. Furthermore, we used the models to predict regulatory mechanisms of DUSPs, where these mechanisms were partially known. Conclusions Boolean modelling is a powerful technique to investigate and understand signalling pathways. We obtained an understanding of different regulatory pathways in breast cancer and new insights on how these signalling pathways are activated. This method can be generalized to other drugs and longer time courses to better understand how resistance to drugs develops in cancer cells over time. Electronic supplementary material The online version of this article (10.1186/s12918-018-0534-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Petronela Buiga
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel.,School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Lydia Tabernero
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jean-Marc Schwartz
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
27
|
Hjort MA, Hov H, Abdollahi P, Vandsemb EN, Fagerli UM, Lund B, Slørdahl TS, Børset M, Rø TB. Phosphatase of regenerating liver-3 (PRL-3) is overexpressed in classical Hodgkin lymphoma and promotes survival and migration. Exp Hematol Oncol 2018; 7:8. [PMID: 29651360 PMCID: PMC5894150 DOI: 10.1186/s40164-018-0100-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/31/2018] [Indexed: 11/25/2022] Open
Abstract
Background Phosphatase of regenerating liver-3 (PRL-3) is implicated in oncogenesis of hematological and solid cancers. PRL-3 expression increases metastatic potential, invasiveness and is associated with poor prognosis. With this study, we aimed to show a possible oncogenic role of PRL-3 in classical Hodgkin lymphoma (cHL). Methods PRL-3 expression was measured in 25 cHL patients by immunohistochemistry and gene expression was analyzed from microdissected malignant cells. We knocked down PRL-3 in the cHL cell lines L1236 and HDLM2 and used small molecular inhibitors against PRL-3 to investigate proliferation, migration and cytokine production. Results PRL-3 protein was expressed in 16% of patient samples. In three different gene expression datasets, PRL-3 was significantly overexpressed compared to normal controls. PRL-3 knockdown reduced proliferation, viability and Mcl-1 expression in L1236, but not in HDLM2 cells. Thienopyridone, a small molecule inhibitor of PRL-3, reduced proliferation of both L1236 and HDLM2. PRL-3 affected IL-13 secretion and enhanced STAT6 signaling. IL-13 stimulation partially rescued proliferation in L1236 cells after knockdown of PRL-3. PRL-3 knockdown reduced migration in both L1236 and HDLM2 cells. Conclusion PRL-3 was overexpressed in a subset of cHL patients. Inhibition of PRL-3 increased IL-13 cytokine production and reduced migration, proliferation and viability. The effects could be mediated through regulation of the anti-apoptotic molecule Mcl-1 and a feedback loop of IL-13 mediated activation of STAT6. This point to a role for PRL-3 in the pathogenesis of Hodgkin lymphoma, and PRL-3 could be a possible new drug target. Electronic supplementary material The online version of this article (10.1186/s40164-018-0100-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Magnus Aassved Hjort
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,2Children's Clinic, Trondheim University Hospital, Trondheim, Norway
| | - Håkon Hov
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,3Department of Pathology, Trondheim University Hospital, Trondheim, Norway
| | - Pegah Abdollahi
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,2Children's Clinic, Trondheim University Hospital, Trondheim, Norway
| | - Esten Nymoen Vandsemb
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,2Children's Clinic, Trondheim University Hospital, Trondheim, Norway
| | - Unn-Merete Fagerli
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,4Cancer Clinic, Trondheim University Hospital, Trondheim, Norway
| | - Bendik Lund
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,2Children's Clinic, Trondheim University Hospital, Trondheim, Norway
| | - Tobias Schmidt Slørdahl
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,5Department of Hematology, Trondheim University Hospital, Trondheim, Norway
| | - Magne Børset
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,6Department of Immunology and Transfusion Medicine St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Torstein Baade Rø
- 1Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, (NTNU), P.O. Box 8905, 7491 Trondheim, Norway.,2Children's Clinic, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
28
|
Morro A, Canals V, Oliver A, Alomar ML, Galan-Prado F, Ballester PJ, Rossello JL. A Stochastic Spiking Neural Network for Virtual Screening. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2018; 29:1371-1375. [PMID: 28186913 DOI: 10.1109/tnnls.2017.2657601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Virtual screening (VS) has become a key computational tool in early drug design and screening performance is of high relevance due to the large volume of data that must be processed to identify molecules with the sought activity-related pattern. At the same time, the hardware implementations of spiking neural networks (SNNs) arise as an emerging computing technique that can be applied to parallelize processes that normally present a high cost in terms of computing time and power. Consequently, SNN represents an attractive alternative to perform time-consuming processing tasks, such as VS. In this brief, we present a smart stochastic spiking neural architecture that implements the ultrafast shape recognition (USR) algorithm achieving two order of magnitude of speed improvement with respect to USR software implementations. The neural system is implemented in hardware using field-programmable gate arrays allowing a highly parallelized USR implementation. The results show that, due to the high parallelization of the system, millions of compounds can be checked in reasonable times. From these results, we can state that the proposed architecture arises as a feasible methodology to efficiently enhance time-consuming data-mining processes such as 3-D molecular similarity search.
Collapse
|
29
|
Salamoun JM, McQueeney KE, Patil K, Geib SJ, Sharlow ER, Lazo JS, Wipf P. Photooxygenation of an amino-thienopyridone yields a more potent PTP4A3 inhibitor. Org Biomol Chem 2018; 14:6398-402. [PMID: 27291491 DOI: 10.1039/c6ob00946h] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The phosphatase PTP4A3 is an attractive anticancer target, but knowledge of its exact role in cells remains incomplete. A potent, structurally novel inhibitor of the PTP4A family was obtained by photooxygenation of a less active, electron-rich thienopyridone (1). Iminothienopyridinedione 13 displays increased solution stability and is readily obtained by two new synthetic routes that converge in the preparation of 1. The late-stage photooxygenation of 1 to give 13 in high yield highlights the potential of this reaction to modify the structure and properties of a biological lead compound and generate value for expanding the scope of an SAR investigation. Analog 13 should become a valuable tool for further exploration of the role of PTP4A3 in tumor progression.
Collapse
Affiliation(s)
- Joseph M Salamoun
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| | - Kelley E McQueeney
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | - Kalyani Patil
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| | - Steven J Geib
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| | - Elizabeth R Sharlow
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | - John S Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.
| |
Collapse
|
30
|
Hoeger B, Rios P, Berteotti A, Hoermann B, Duan G, Köhn M. Mutational Analysis of a Conserved Glutamate Reveals Unique Mechanistic and Structural Features of the Phosphatase PRL-3. ACS OMEGA 2017; 2:9171-9180. [PMID: 30023603 PMCID: PMC6044973 DOI: 10.1021/acsomega.7b01208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/23/2017] [Indexed: 06/08/2023]
Abstract
Phosphatase of regenerating liver (PRL)-3 (PTP4A3) has gained much attention in cancer research due to its involvement in tumor promoting and metastatic processes. It belongs to the protein tyrosine phosphatase (PTP) superfamily and is thought to follow the catalytic mechanism shared by this family, which aside from the conserved active-site amino acids includes a conserved glutamic acid residue that is usually required for the integrity of the active site in PTPs. We noted that in structures of PRL-3, PRL-1, and PTEN these residues do not clearly align and therefore we sought to investigate if the glutamic acid residue fulfills its usual function in these proteins. Although this residue was essential for PTEN's catalytic activity, it was nonessential for PRL-1 and PRL-3. Surprisingly, the mutation E50R increased PRL-3 activity against all tested in vitro substrates and also enhanced PRL-3-promoted cell adhesion and migration. We show that the introduction of Arg50 leads to an enhancement of substrate turnover for both PRL-3 and, to a lesser extent, PRL-1, and that the stronger gain in activity correlates with a higher structural flexibility of PRL-3, likely allowing for conformational adaptation during catalysis. Thus, in contrast to its crucial functions in other PTPs, this conserved glutamic acid can be replaced in PRL-3 without impairing the structural integrity. The variant with enhanced activity might serve as a tool to study PRL-3 in the future.
Collapse
Affiliation(s)
- Birgit Hoeger
- Genome
Biology Unit, European Molecular Biology
Laboratory, Meyerhofstraße
1, 69117 Heidelberg, Germany
| | - Pablo Rios
- Genome
Biology Unit, European Molecular Biology
Laboratory, Meyerhofstraße
1, 69117 Heidelberg, Germany
- Faculty
of Biology and Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Anna Berteotti
- Genome
Biology Unit, European Molecular Biology
Laboratory, Meyerhofstraße
1, 69117 Heidelberg, Germany
| | - Bernhard Hoermann
- Genome
Biology Unit, European Molecular Biology
Laboratory, Meyerhofstraße
1, 69117 Heidelberg, Germany
- Faculty
of Biology and Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- University
of Heidelberg, 69120 Heidelberg, Germany
| | - Guangyou Duan
- Genome
Biology Unit, European Molecular Biology
Laboratory, Meyerhofstraße
1, 69117 Heidelberg, Germany
| | - Maja Köhn
- Genome
Biology Unit, European Molecular Biology
Laboratory, Meyerhofstraße
1, 69117 Heidelberg, Germany
- Faculty
of Biology and Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| |
Collapse
|
31
|
Andersen S, Richardsen E, Rakaee M, Bertilsson H, Bremnes R, Børset M, Busund LT, Slørdahl T. Expression of phosphatase of regenerating liver (PRL)-3, is independently associated with biochemical failure, clinical failure and death in prostate cancer. PLoS One 2017; 12:e0189000. [PMID: 29190795 PMCID: PMC5708709 DOI: 10.1371/journal.pone.0189000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/16/2017] [Indexed: 11/19/2022] Open
Abstract
Background Prostate cancer (PC) stratification needs new prognostic tools to reduce overtreatment. Phosphatase of regenerating liver (PRL-3) is a phosphatase found at high levels in several cancer types, where its expression is associated with survival. A recent PC cell line study has shown it to be involved in PC growth and migration. Methods We used a monoclonal antibody to evaluate the expression of PRL-3 in PC tissue of patients in an unselected cohort of 535 prostatectomy patients. We analyzed associations between PRL-3 expression and biochemical failure-free survival (BFFS), clinical failure-free survival (CFFS) and PC death-free survival (PCDFS). Results Cytoplasmic PRL-3 staining in tumor cells was significantly correlated to expression of molecules in the VEGFR-axis, but not to the clinicopathological variables. High PRL-3 was not significantly associated with survival in the univariate analysis for BFFS (p = 0.131), but significantly associated with CFFS (p = 0.044) and PCDFS (p = 0.041). In multivariate analysis for the various end points, PRL-3 came out as an independent and significant indicator of poor survival for BFFS (HR = 1.53, CI95% 1.10–2.13, p = 0.012), CFFS (HR = 2.41, CI95% 1.17–4.98, p = 0.017) and PCDFS (HR = 3.99, CI95% 1.21–13.1, p = 0.023). Conclusions PRL-3 is independently associated with all PC endpoints in this study. Since high PRL-3 expression also correlates with poor prognosis in other cancers and functional studies in PC support these findings, PRL-3 emerges as a potential treatment target in PC.
Collapse
Affiliation(s)
- Sigve Andersen
- Translational Cancer Research Group, Department Clinical Medicine, UiT, The Arctic University of Norway, Tromso, Norway
- Department Oncology, University Hospital of North Norway, Tromso, Norway
- * E-mail:
| | - Elin Richardsen
- Translational Cancer Research Group, Department of Medical Biology, UiT, The Arctic University of Norway, Tromso, Norway
- Department Pathology, University Hospital of North Norway, Tromso, Norway
| | - Mehrdad Rakaee
- Translational Cancer Research Group, Department of Medical Biology, UiT, The Arctic University of Norway, Tromso, Norway
| | - Helena Bertilsson
- Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Urology, St. Olavs Hospital - Trondheim University Hospital, Trondheim, Norway
| | - Roy Bremnes
- Translational Cancer Research Group, Department Clinical Medicine, UiT, The Arctic University of Norway, Tromso, Norway
- Department Oncology, University Hospital of North Norway, Tromso, Norway
| | - Magne Børset
- Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital - Trondheim University Hospital, Trondheim, Norway
| | - Lill-Tove Busund
- Translational Cancer Research Group, Department of Medical Biology, UiT, The Arctic University of Norway, Tromso, Norway
- Department Pathology, University Hospital of North Norway, Tromso, Norway
| | - Tobias Slørdahl
- Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Hematology, St. Olavs Hospital - Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
32
|
Small molecule targeting of PTPs in cancer. Int J Biochem Cell Biol 2017; 96:171-181. [PMID: 28943273 DOI: 10.1016/j.biocel.2017.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 01/28/2023]
Abstract
Protein tyrosine phosphatases (PTPs) undeniably have a central role in the development and progression of human cancers. Historically, however, PTPs have not been viewed as privileged drug targets, and progress on identifying potent, selective, and cell-active small molecule PTP inhibitors has suffered accordingly. This situation is rapidly changing, however, due to biochemical advances in the study of PTPs and recent small molecule screening campaigns, which have identified potent and mechanistically diverse lead structures. These compounds are facilitating the exploration of the fundamental cellular processes controlled by PTPs in cancers, and could form the inflection point for new therapeutic paradigms for the treatment of a range of cancers. Herein, we review recent advances in the discovery and biological annotation of cancer-relevant small molecule PTP inhibitors.
Collapse
|
33
|
Bhore N, Wang BJ, Chen YW, Liao YF. Critical Roles of Dual-Specificity Phosphatases in Neuronal Proteostasis and Neurological Diseases. Int J Mol Sci 2017; 18:ijms18091963. [PMID: 28902166 PMCID: PMC5618612 DOI: 10.3390/ijms18091963] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 12/31/2022] Open
Abstract
Protein homeostasis or proteostasis is a fundamental cellular property that encompasses the dynamic balancing of processes in the proteostasis network (PN). Such processes include protein synthesis, folding, and degradation in both non-stressed and stressful conditions. The role of the PN in neurodegenerative disease is well-documented, where it is known to respond to changes in protein folding states or toxic gain-of-function protein aggregation. Dual-specificity phosphatases have recently emerged as important participants in maintaining balance within the PN, acting through modulation of cellular signaling pathways that are involved in neurodegeneration. In this review, we will summarize recent findings describing the roles of dual-specificity phosphatases in neurodegeneration and offer perspectives on future therapeutic directions.
Collapse
Affiliation(s)
- Noopur Bhore
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Bo-Jeng Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Yun-Wen Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Yung-Feng Liao
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
34
|
Frankson R, Yu ZH, Bai Y, Li Q, Zhang RY, Zhang ZY. Therapeutic Targeting of Oncogenic Tyrosine Phosphatases. Cancer Res 2017; 77:5701-5705. [PMID: 28855209 DOI: 10.1158/0008-5472.can-17-1510] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/12/2017] [Accepted: 08/24/2017] [Indexed: 01/01/2023]
Abstract
Protein tyrosine phosphatases (PTP) are exciting and novel targets for cancer drug discovery that work in concert with protein tyrosine kinases (PTK) in controlling cellular homeostasis. Given the activating role that some PTKs play in initiating growth factor-mediated cellular processes, PTPs are usually perceived as the negative regulators of these events and therefore tumor suppressive in nature. However, mounting evidence indicate that PTPs do not always antagonize the activity of PTKs in regulating tyrosine phosphorylation, but can also play dominant roles in the initiation and progression of signaling cascades that regulate cell functions. It follows, therefore, that PTP malfunction can actively contribute to a host of human disorders, in particular, cancer, metabolic syndromes, and autoimmune diseases. The Src homology domain containing phosphatase 2 (SHP2) and the three-membered family of phosphatases of regenerating liver (PRL) are infamously oncogenic members of the PTP superfamily. Both are established regulators of major cancer pathways such as Ras/ERK1/2, Src, JAK/STAT, JNK, NF-κB, and PTEN/PI3K/AKT. Furthermore, upregulation, mutation, or other dysregulation of these PTPs has been positively correlated with cancer initiation and progression. This review will provide topical coverage of target validation and drug discovery efforts made in targeting these oncogenic PTPs as compelling candidates for cancer therapy. Cancer Res; 77(21); 5701-5. ©2017 AACR.
Collapse
Affiliation(s)
- Rochelle Frankson
- Departments of Medicinal Chemistry and Molecular Pharmacology and Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
| | - Zhi-Hong Yu
- Departments of Medicinal Chemistry and Molecular Pharmacology and Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
| | - Yunpeng Bai
- Departments of Medicinal Chemistry and Molecular Pharmacology and Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
| | - Qinglin Li
- Departments of Medicinal Chemistry and Molecular Pharmacology and Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
| | - Ruo-Yu Zhang
- Departments of Medicinal Chemistry and Molecular Pharmacology and Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
| | - Zhong-Yin Zhang
- Departments of Medicinal Chemistry and Molecular Pharmacology and Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana.
| |
Collapse
|
35
|
Lazo JS, McQueeney KE, Sharlow ER. New Approaches to Difficult Drug Targets: The Phosphatase Story. SLAS DISCOVERY 2017; 22:1071-1083. [DOI: 10.1177/2472555217721142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The drug discovery landscape is littered with promising therapeutic targets that have been abandoned because of insufficient validation, historical screening failures, and inferior chemotypes. Molecular targets once labeled as “undruggable” or “intractable” are now being more carefully interrogated, and while they remain challenging, many target classes are appearing more approachable. Protein tyrosine phosphatases represent an excellent example of a category of molecular targets that have emerged as druggable, with several small molecules and antibodies recently becoming available for further development. In this review, we examine some of the diseases that are associated with protein tyrosine phosphatase dysfunction and use some prototype contemporary strategies to illustrate approaches that are being used to identify small molecules targeting this enzyme class.
Collapse
Affiliation(s)
- John S. Lazo
- Department of Pharmacology, Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, VA, USA
| | - Kelley E. McQueeney
- Department of Pharmacology, Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, VA, USA
| | - Elizabeth R. Sharlow
- Department of Pharmacology, Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
36
|
Oliver A, Canals V, Rosselló JL. A Bayesian Target Predictor Method based on Molecular Pairing Energies estimation. Sci Rep 2017; 7:43738. [PMID: 28263323 PMCID: PMC5338323 DOI: 10.1038/srep43738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/30/2017] [Indexed: 11/13/2022] Open
Abstract
Virtual screening (VS) is applied in the early drug discovery phases for the quick inspection of huge molecular databases to identify those compounds that most likely bind to a given drug target. In this context, there is the necessity of the use of compact molecular models for database screening and precise target prediction in reasonable times. In this work we present a new compact energy-based model that is tested for its application to Virtual Screening and target prediction. The model can be used to quickly identify active compounds in huge databases based on the estimation of the molecule’s pairing energies. The greatest molecular polar regions along with its geometrical distribution are considered by using a short set of smart energy vectors. The model is tested using similarity searches within the Directory of Useful Decoys (DUD) database. The results obtained are considerably better than previously published models. As a Target prediction methodology we propose the use of a Bayesian Classifier that uses a combination of different active compounds to build an energy-dependent probability distribution function for each target.
Collapse
Affiliation(s)
- Antoni Oliver
- Physics Department, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Vincent Canals
- Physics Department, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Josep L Rosselló
- Physics Department, Universitat de les Illes Balears, Palma de Mallorca, Spain
| |
Collapse
|
37
|
Abstract
Phosphatases play key roles in normal physiology and diseases. Studying phosphatases has been both essential and challenging, and the application of conventional genetic and biochemical methods has led to crucial but still limited understanding of their mechanisms, substrates, and exclusive functions within highly intricate networks. With the advances in technologies such as cellular imaging and molecular and chemical biology in terms of sensitive tools and methods, the phosphatase field has thrived in the past years and has set new insights for cell signaling studies and for therapeutic development. In this review, we give an overview of the existing interdisciplinary tools for phosphatases, give examples on how they have been applied to increase our understanding of these enzymes, and suggest how they-and other tools yet barely used in the phosphatase field-might be adapted to address future questions and challenges.
Collapse
Affiliation(s)
- Sara Fahs
- European Molecular Biology Laboratory, Genome Biology
Unit, Meyerhofstrasse
1, 69117 Heidelberg, Germany
| | - Pablo Lujan
- European Molecular Biology Laboratory, Genome Biology
Unit, Meyerhofstrasse
1, 69117 Heidelberg, Germany
| | - Maja Köhn
- European Molecular Biology Laboratory, Genome Biology
Unit, Meyerhofstrasse
1, 69117 Heidelberg, Germany
| |
Collapse
|
38
|
Luján P, Varsano G, Rubio T, Hennrich ML, Sachsenheimer T, Gálvez-Santisteban M, Martín-Belmonte F, Gavin AC, Brügger B, Köhn M. PRL-3 disrupts epithelial architecture by altering the post-mitotic midbody position. J Cell Sci 2016; 129:4130-4142. [PMID: 27656108 PMCID: PMC5117205 DOI: 10.1242/jcs.190215] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022] Open
Abstract
Disruption of epithelial architecture is a fundamental event during epithelial tumorigenesis. We show that the expression of the cancer-promoting phosphatase PRL-3 (PTP4A3), which is overexpressed in several epithelial cancers, in polarized epithelial MDCK and Caco2 cells leads to invasion and the formation of multiple ectopic, fully polarized lumens in cysts. Both processes disrupt epithelial architecture and are hallmarks of cancer. The pathological relevance of these findings is supported by the knockdown of endogenous PRL-3 in MCF-7 breast cancer cells grown in three-dimensional branched structures, showing the rescue from multiple-lumen- to single-lumen-containing branch ends. Mechanistically, it has been previously shown that ectopic lumens can arise from midbodies that have been mislocalized through the loss of mitotic spindle orientation or through the loss of asymmetric abscission. Here, we show that PRL-3 triggers ectopic lumen formation through midbody mispositioning without altering the spindle orientation or asymmetric abscission, instead, PRL-3 accelerates cytokinesis, suggesting that this process is an alternative new mechanism for ectopic lumen formation in MDCK cysts. The disruption of epithelial architecture by PRL-3 revealed here is a newly recognized mechanism for PRL-3-promoted cancer progression.
Collapse
Affiliation(s)
- Pablo Luján
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Giulia Varsano
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Teresa Rubio
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Marco L Hennrich
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg 69117, Germany
| | - Timo Sachsenheimer
- Heidelberg University Biochemistry Center, University of Heidelberg, Heidelberg 69120, Germany
| | - Manuel Gálvez-Santisteban
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28049, Spain
| | - Fernando Martín-Belmonte
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28049, Spain
| | - Anne-Claude Gavin
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg 69117, Germany
| | - Britta Brügger
- Heidelberg University Biochemistry Center, University of Heidelberg, Heidelberg 69120, Germany
| | - Maja Köhn
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| |
Collapse
|
39
|
Li H, Leung KS, Wong MH, Ballester PJ. USR-VS: a web server for large-scale prospective virtual screening using ultrafast shape recognition techniques. Nucleic Acids Res 2016; 44:W436-41. [PMID: 27106057 PMCID: PMC4987897 DOI: 10.1093/nar/gkw320] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/06/2016] [Indexed: 12/12/2022] Open
Abstract
Ligand-based Virtual Screening (VS) methods aim at identifying molecules with a similar activity profile across phenotypic and macromolecular targets to that of a query molecule used as search template. VS using 3D similarity methods have the advantage of biasing this search toward active molecules with innovative chemical scaffolds, which are highly sought after in drug design to provide novel leads with improved properties over the query molecule (e.g. patentable, of lower toxicity or increased potency). Ultrafast Shape Recognition (USR) has demonstrated excellent performance in the discovery of molecules with previously-unknown phenotypic or target activity, with retrospective studies suggesting that its pharmacophoric extension (USRCAT) should obtain even better hit rates once it is used prospectively. Here we present USR-VS (http://usr.marseille.inserm.fr/), the first web server using these two validated ligand-based 3D methods for large-scale prospective VS. In about 2 s, 93.9 million 3D conformers, expanded from 23.1 million purchasable molecules, are screened and the 100 most similar molecules among them in terms of 3D shape and pharmacophoric properties are shown. USR-VS functionality also provides interactive visualization of the similarity of the query molecule against the hit molecules as well as vendor information to purchase selected hits in order to be experimentally tested.
Collapse
Affiliation(s)
- Hongjian Li
- Institute of Future Cities, Chinese University of Hong Kong, Hong Kong
| | - Kwong-S Leung
- Department of Computer Science and Engineering, Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong
| | - Man-H Wong
- Department of Computer Science and Engineering, Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong
| | - Pedro J Ballester
- Cancer Research Center of Marseille, INSERM U1068, 13009-Marseille, France
| |
Collapse
|
40
|
Kostantin E, Hardy S, Valinsky WC, Kompatscher A, de Baaij JHF, Zolotarov Y, Landry M, Uetani N, Martínez-Cruz LA, Hoenderop JGJ, Shrier A, Tremblay ML. Inhibition of PRL-2·CNNM3 Protein Complex Formation Decreases Breast Cancer Proliferation and Tumor Growth. J Biol Chem 2016; 291:10716-25. [PMID: 26969161 PMCID: PMC4865918 DOI: 10.1074/jbc.m115.705863] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 11/06/2022] Open
Abstract
The oncogenic phosphatase of regenerating liver 2 (PRL-2) has been shown to regulate intracellular magnesium levels by forming a complex through an extended amino acid loop present in the Bateman module of the CNNM3 magnesium transporter. Here we identified highly conserved residues located on this amino acid loop critical for the binding with PRL-2. A single point mutation (D426A) of one of those critical amino acids was found to completely disrupt PRL-2·human Cyclin M 3 (CNNM3) complex formation. Whole-cell voltage clamping revealed that expression of CNNM3 influenced the surface current, whereas overexpression of the binding mutant had no effect, indicating that the binding of PRL-2 to CNNM3 is important for the activity of the complex. Interestingly, overexpression of the CNNM3 D426A-binding mutant in cancer cells decreased their ability to proliferate under magnesium-deprived situations and under anchorage-independent growth conditions, demonstrating a PRL-2·CNNM3 complex-dependent oncogenic advantage in a more stringent environment. We further confirmed the importance of this complex in vivo using an orthotopic xenograft breast cancer model. Finally, because molecular modeling showed that the Asp-426 side chain in CNNM3 buries into the catalytic cavity of PRL-2, we showed that a PRL inhibitor could abrogate complex formation, resulting in a decrease in proliferation of human breast cancer cells. In summary, we provide evidence that this fundamental regulatory aspect of PRL-2 in cancer cells could potentially lead to broadly applicable and innovative therapeutic avenues.
Collapse
Affiliation(s)
- Elie Kostantin
- From the Rosalind and Morris Goodman Cancer Research Centre, Montréal, Québec H3A 1A3, Canada, the Departments of Biochemistry and
| | - Serge Hardy
- From the Rosalind and Morris Goodman Cancer Research Centre, Montréal, Québec H3A 1A3, Canada
| | | | - Andreas Kompatscher
- the Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands, and
| | - Jeroen H F de Baaij
- the Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands, and
| | - Yevgen Zolotarov
- From the Rosalind and Morris Goodman Cancer Research Centre, Montréal, Québec H3A 1A3, Canada, the Departments of Biochemistry and
| | - Melissa Landry
- From the Rosalind and Morris Goodman Cancer Research Centre, Montréal, Québec H3A 1A3, Canada
| | - Noriko Uetani
- From the Rosalind and Morris Goodman Cancer Research Centre, Montréal, Québec H3A 1A3, Canada
| | - Luis Alfonso Martínez-Cruz
- the Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Joost G J Hoenderop
- the Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands, and
| | - Alvin Shrier
- Physiology, McGill University, Montréal, Québec H3A 0G4, Canada
| | - Michel L Tremblay
- From the Rosalind and Morris Goodman Cancer Research Centre, Montréal, Québec H3A 1A3, Canada, the Departments of Biochemistry and
| |
Collapse
|
41
|
Stadlbauer S, Rios P, Ohmori K, Suzuki K, Köhn M. Procyanidins Negatively Affect the Activity of the Phosphatases of Regenerating Liver. PLoS One 2015; 10:e0134336. [PMID: 26226290 PMCID: PMC4520450 DOI: 10.1371/journal.pone.0134336] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 07/08/2015] [Indexed: 01/10/2023] Open
Abstract
Natural polyphenols like oligomeric catechins (procyanidins) derived from green tea and herbal medicines are interesting compounds for pharmaceutical research due to their ability to protect against carcinogenesis in animal models. It is nevertheless still unclear how intracellular pathways are modulated by polyphenols. Monomeric polyphenols were shown to affect the activity of some protein phosphatases (PPs). The three phosphatases of regenerating liver (PRLs) are close relatives and promising therapeutic targets in cancer. In the present study we show that several procyanidins inhibit the activity of all three members of the PRL family in the low micromolar range, whereas monomeric epicatechins show weak inhibitory activity. Increasing the number of catechin units in procyanidins to more than three does not further enhance the potency. Remarkably, the tested procyanidins showed selectivity in vitro when compared to other PPs, and over 10-fold selectivity toward PRL-1 over PRL-2 and PRL-3. As PRL overexpression induces cell migration compared to control cells, the effect of procyanidins on this phenotype was studied. Treatment with procyanidin C2 led to a decrease in cell migration of PRL-1- and PRL-3-overexpressing cells, suggesting the compound-dependent inhibition of PRL-promoted cell migration. Treatment with procyanidin B3 led to selective suppression of PRL-1 overexpressing cells, thereby corroborating the selectivity toward PRL-1- over PRL-3 in vitro. Together, our results show that procyanidins negatively affect PRL activity, suggesting that PRLs could be targets in the polypharmacology of natural polyphenols. Furthermore, they are interesting candidates for the development of PRL-1 inhibitors due to their low cellular toxicity and the selectivity within the PRL family.
Collapse
Affiliation(s)
- Sven Stadlbauer
- European Molecular Biology Laboratory, Genome Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
- * E-mail: (SS); (MK)
| | - Pablo Rios
- European Molecular Biology Laboratory, Genome Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Ken Ohmori
- Tokyo Institute of Technology, Department of Chemistry, O-okayama, Meguro-ku, Tokyo, 152–8551, Japan
| | - Keisuke Suzuki
- Tokyo Institute of Technology, Department of Chemistry, O-okayama, Meguro-ku, Tokyo, 152–8551, Japan
| | - Maja Köhn
- European Molecular Biology Laboratory, Genome Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
- * E-mail: (SS); (MK)
| |
Collapse
|
42
|
Jones LH, Heinis C. Chemical biology & drug discovery. Eur J Med Chem 2014; 88:1-2. [PMID: 25307206 DOI: 10.1016/j.ejmech.2014.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Lyn H Jones
- Worldwide Medicinal Chemistry, Pfizer BioTherapeutics Chemistry, Cambridge, MA, USA.
| | - Christian Heinis
- Laboratory of Therapeutic Proteins and Peptides (LPPT), École Polytechnique Fédérale de Lausanne (EPFL), BCH 5305 (Bâtochime), Lausanne CH-1015, Switzerland.
| |
Collapse
|