1
|
Chun SY, Park C, Oh J, Yoon HJ, Kim TI, Kim Y, Ham SW, Koh HR, Lee HH, Kim HY, Oh K. (Thio)chromenone derivatives exhibit anti-metastatic effects through selective inhibition of uPAR in cancer cell lines: discovery of an uPAR-targeting fluorescent probe. Chem Commun (Camb) 2025; 61:909-912. [PMID: 39668665 DOI: 10.1039/d4cc05907g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
A class of (thio)chromenone derivatives has been identified as suitable ligands for uPAR, a glycoprotein with a prognostic value in a large number of human cancers. The (thio)chromenone agents actively inhibited the binding of uPAR to uPA with a binding affinity of 18.6 nM, reducing cell migration in the wound healing assay by up to 40% without apparent cell motility. The discovery of an uPAR-targeting fluorescent probe was also made in this study that can selectively bind to the membrane uPAR, providing valuable molecular insights into the role of uPAR in cancer metastasis. This study should serve as a basis for the development of new uPAR-targeting agents that can control the metastatic potential of cancer cells with minimal cytotoxicity.
Collapse
Affiliation(s)
- So-Young Chun
- Center for Metareceptome Research, Graduate School of Pharmaceutical Sciences, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea.
| | - Chanhee Park
- Center for Metareceptome Research, Graduate School of Pharmaceutical Sciences, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea.
| | - Jiwon Oh
- Department of Integrative Energy Engineering, Graduate School of Energy and Environment (KU-KIST Green School), College of Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hey-Jin Yoon
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Tae-Il Kim
- Department of Chemistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Youngmi Kim
- Department of Chemistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seung Wook Ham
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea
| | - Hye Ran Koh
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hun Young Kim
- Department of Global Innovative Drugs, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea.
| | - Kyungsoo Oh
- Center for Metareceptome Research, Graduate School of Pharmaceutical Sciences, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea.
| |
Collapse
|
2
|
Bum-Erdene K, Yeh IJ, Gonzalez-Gutierrez G, Ghozayel MK, Pollok K, Meroueh SO. Small-Molecule Cyanamide Pan-TEAD·YAP1 Covalent Antagonists. J Med Chem 2023; 66:266-284. [PMID: 36562717 DOI: 10.1021/acs.jmedchem.2c01189] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transcriptional enhanced associate domains (TEADs) are transcription factors that bind to cotranscriptional activators like the yes-associated protein (YAP) or its paralog transcriptional coactivator with a PDZ-binding motif (TAZ). TEAD·YAP/TAZ target genes are involved in tissue and immune homeostasis, organ size control, tumor growth, and metastasis. Here, we report isoindoline and octahydroisoindole small molecules with a cyanamide electrophile that forms a covalent bond with a conserved cysteine in the TEAD palmitate-binding cavity. Time- and concentration-dependent studies against TEAD1-4 yielded second-order rate constants kinact/KI greater than 100 M-1 s-1. Compounds inhibited YAP1 binding to TEADs with submicromolar IC50 values. Cocrystal structures with TEAD2 enabled structure-activity relationship studies. In mammalian cells, compounds suppressed CTGF mRNA levels and inhibited TEAD1-4 transcriptional activity with submicromolar IC50 values. Inhibition of TEAD binding to YAP1 in mammalian cells was also observed. Several compounds inhibited the cell viability of sarcoma, hepatocellular carcinoma, glioblastoma, and breast cancer cells with single-digit micromolar IC50 values.
Collapse
Affiliation(s)
- Khuchtumur Bum-Erdene
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - I-Ju Yeh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Giovanni Gonzalez-Gutierrez
- Department of Molecular and Cellular Biochemistry, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - Mona K Ghozayel
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Karen Pollok
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| |
Collapse
|
3
|
Arancillo M, Lin CM, Burgess K. Piptide Chemotypes for Perturbation of the Interaction of Urokinase with Its Receptor. J Med Chem 2022; 65:12925-12932. [PMID: 36166370 DOI: 10.1021/acs.jmedchem.2c00759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Only a few small molecules that disrupt the uPA and uPA receptor (uPAR) interaction have been discovered despite decades of research in the area, and none have been approved in clinical trials. Research reported here features two new ways of considering the problem of discovering small molecules to disrupt uPA•uPAR, specifically in terms of chemotype design and method of evaluation. Chemotypes used in this work are piptides (Arancillo . Angew. Chem., Int. Ed., 2021, 60, 6653-6659) with side chains corresponding to the uPA loop that binds uPAR. Further, hybrids of 1 and another uPAR ligand developed in these labs (2), i.e., 3 and 4, were also designed and tested. All the piptide chemotypes bound uPAR at concentrations of 50 μM or less. Members of this series had Ki values <3 μM and showed favorable responses in cellular assays; these data are comparable with the best small molecule uPA•uPAR disruptors in the literature (from conventional screening).
Collapse
Affiliation(s)
- Maritess Arancillo
- Department of Chemistry, Texas A&M University, P.O. Box 30012, College Station, Texas 77842, United States
| | - Chen-Ming Lin
- Department of Chemistry, Texas A&M University, P.O. Box 30012, College Station, Texas 77842, United States
| | - Kevin Burgess
- Department of Chemistry, Texas A&M University, P.O. Box 30012, College Station, Texas 77842, United States
| |
Collapse
|
4
|
Zhai BT, Tian H, Sun J, Zou JB, Zhang XF, Cheng JX, Shi YJ, Fan Y, Guo DY. Urokinase-type plasminogen activator receptor (uPAR) as a therapeutic target in cancer. J Transl Med 2022; 20:135. [PMID: 35303878 PMCID: PMC8932206 DOI: 10.1186/s12967-022-03329-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/03/2022] [Indexed: 12/22/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR) is an attractive target for the treatment of cancer, because it is expressed at low levels in healthy tissues but at high levels in malignant tumours. uPAR is closely related to the invasion and metastasis of malignant tumours, plays important roles in the degradation of extracellular matrix (ECM), tumour angiogenesis, cell proliferation and apoptosis, and is associated with the multidrug resistance (MDR) of tumour cells, which has important guiding significance for the judgement of tumor malignancy and prognosis. Several uPAR-targeted antitumour therapeutic agents have been developed to suppress tumour growth, metastatic processes and drug resistance. Here, we review the recent advances in the development of uPAR-targeted antitumor therapeutic strategies, including nanoplatforms carrying therapeutic agents, photodynamic therapy (PDT)/photothermal therapy (PTT) platforms, oncolytic virotherapy, gene therapy technologies, monoclonal antibody therapy and tumour immunotherapy, to promote the translation of these therapeutic agents to clinical applications.
Collapse
Affiliation(s)
- Bing-Tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-Bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-Fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-Xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Ya-Jun Shi
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yu Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-Yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| |
Collapse
|
5
|
Passirani C, Vessières A, La Regina G, Link W, Silvestri R. Modulating undruggable targets to overcome cancer therapy resistance. Drug Resist Updat 2021; 60:100788. [DOI: 10.1016/j.drup.2021.100788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/03/2022]
|
6
|
Leth JM, Ploug M. Targeting the Urokinase-Type Plasminogen Activator Receptor (uPAR) in Human Diseases With a View to Non-invasive Imaging and Therapeutic Intervention. Front Cell Dev Biol 2021; 9:732015. [PMID: 34490277 PMCID: PMC8417595 DOI: 10.3389/fcell.2021.732015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022] Open
Abstract
The interaction between the serine protease urokinase-type plasminogen activator (uPA) and its glycolipid-anchored receptor (uPAR) focalizes plasminogen activation to cell surfaces, thereby regulating extravascular fibrinolysis, cell adhesion, and migration. uPAR belongs to the Ly6/uPAR (LU) gene superfamily and the high-affinity binding site for uPA is assembled by a dynamic association of its three consecutive LU domains. In most human solid cancers, uPAR is expressed at the invasive areas of the tumor-stromal microenvironment. High levels of uPAR in resected tumors or shed to the plasma of cancer patients are robustly associated with poor prognosis and increased risk of relapse and metastasis. Over the years, a plethora of different strategies to inhibit uPA and uPAR function have been designed and investigated in vitro and in vivo in mouse models, but so far none have been implemented in the clinics. In recent years, uPAR-targeting with the intent of cytotoxic eradication of uPAR-expressing cells have nonetheless gained increasing momentum. Another avenue that is currently being explored is non-invasive imaging with specific uPAR-targeted reporter-molecules containing positron emitting radionuclides or near-infrared (NIR) florescence probes with the overarching aim of being able to: (i) localize disease dissemination using positron emission tomography (PET) and (ii) assist fluorescence guided surgery using optical imaging. In this review, we will discuss these advancements with special emphasis on applications using a small 9-mer peptide antagonist that targets uPAR with high affinity.
Collapse
Affiliation(s)
- Julie Maja Leth
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Bum-Erdene K, Liu D, Xu D, Ghozayel MK, Meroueh SO. Design and Synthesis of Fragment Derivatives with a Unique Inhibition Mechanism of the uPAR·uPA Interaction. ACS Med Chem Lett 2021; 12:60-66. [PMID: 33488965 DOI: 10.1021/acsmedchemlett.0c00422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
There is substantial interest in the development of small molecules that inhibit the tight and highly challenging protein-protein interaction between the glycophosphatidylinositol (GPI)-anchored cell surface receptor uPAR and the serine protease uPA. While preparing derivatives of a fragment-like compound that previously emerged from a computational screen, we identified compound 5 (IPR-3242), which inhibited binding of uPA to uPAR with submicromolar IC50s. The high inhibition potency prompted us to carry out studies to rule out potential aggregation, lack of stability, reactivity, and nonspecific inhibition. We designed and prepared 16 derivatives to further explore the role of each substituent. Interestingly, the compounds only partially inhibited binding of a fluorescently labeled α-helical peptide that binds to uPAR at the uPAR·uPA interface. Collectively, the results suggest that the compounds bind to uPAR outside of the uPAR·uPA interface, trapping the receptor into a conformation that is not able to bind to uPA. Additional studies will have to be carried out to determine whether this unique inhibition mechanism can occur at the cell surface.
Collapse
Affiliation(s)
- Khuchtumur Bum-Erdene
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Degang Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - David Xu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Mona K. Ghozayel
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Samy O. Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| |
Collapse
|
8
|
Xu D, Bum-Erdene K, Leth JM, Ghozayel MK, Ploug M, Meroueh SO. Small-Molecule Inhibition of the uPAR ⋅ uPA Interaction by Conformational Selection. ChemMedChem 2020; 16:377-387. [PMID: 33107192 DOI: 10.1002/cmdc.202000558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/25/2020] [Indexed: 12/12/2022]
Abstract
The urokinase receptor (uPAR) is a cell surface receptor that binds to the serine protease urokinase-type plasminogen activator (uPA) with high affinity. This interaction is beneficial for extravascular fibrin clearance, but it has also been associated with a broad range of pathological conditions including cancer, atherosclerosis, and kidney disease. Here, starting with a small molecule that we previously discovered by virtual screening and cheminformatics analysis, we design and synthesize several derivatives that were tested for binding and inhibition of the uPAR ⋅ uPA interaction. To confirm the binding site and establish a binding mode of the compounds, we carried out biophysical studies using uPAR mutants, among them uPARH47C-N259C , a mutant previously developed to mimic the structure of uPA-bound uPAR. Remarkably, a substantial increase in potency is observed for inhibition of uPARH47C-N259C binding to uPA compared to wild-type uPAR, consistent with our use of the structure of uPAR in its uPA-bound state to design small-molecule uPAR ⋅ uPA antagonists. Combined with the biophysical studies, molecular docking followed by extensive explicit-solvent molecular dynamics simulations and MM-GBSA free energy calculations yielded the most favorable binding pose of the compound. Collectively, these results suggest that potent inhibition of uPAR binding to uPA with small molecules will likely only be achieved by developing small molecules that exhibit high-affinity to solution apo structures of uPAR, rather than uPA-bound structures of the receptor.
Collapse
Affiliation(s)
- David Xu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Khuchtumur Bum-Erdene
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Julie M Leth
- Finsen Laboratory, Rigshospitalet, 2200, Copenhagen N, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Mona K Ghozayel
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, 2200, Copenhagen N, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Lin C, Arancillo M, Whisenant J, Burgess K. Unconventional Secondary Structure Mimics: Ladder‐Rungs. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Chen‐Ming Lin
- Department of Chemistry Texas A&M University Box 30012 College Station TX 77842 USA
| | - Maritess Arancillo
- Department of Chemistry Texas A&M University Box 30012 College Station TX 77842 USA
| | - Jonathan Whisenant
- Department of Chemistry Texas A&M University Box 30012 College Station TX 77842 USA
| | - Kevin Burgess
- Department of Chemistry Texas A&M University Box 30012 College Station TX 77842 USA
| |
Collapse
|
10
|
Lin CM, Arancillo M, Whisenant J, Burgess K. Unconventional Secondary Structure Mimics: Ladder-Rungs. Angew Chem Int Ed Engl 2020; 59:9398-9402. [PMID: 32176815 DOI: 10.1002/anie.202002639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/06/2020] [Indexed: 01/31/2023]
Abstract
Secondary structures tend to be recognizable because they have repeating structural motifs, but mimicry of these does not have to follow such well-defined patterns. Bioinformatics studies to match side-chain orientations of a novel hydantoin triazole chemotype (1) to protein-protein interfaces revealed it tends to align well across parallel and antiparallel sheets, like rungs on a ladder. One set of these overlays was observed for the protein-protein interaction uPA⋅uPAR. Consequently, chemotype 1 was made with appropriate side-chains to mimic uPA at this interface. Biophysical assays indicate these compounds did in fact bind uPAR, and elicit cellular responses that affected invasion, migration, and wound healing.
Collapse
Affiliation(s)
- Chen-Ming Lin
- Department of Chemistry, Texas A&M University, Box 30012, College Station, TX, 77842, USA
| | - Maritess Arancillo
- Department of Chemistry, Texas A&M University, Box 30012, College Station, TX, 77842, USA
| | - Jonathan Whisenant
- Department of Chemistry, Texas A&M University, Box 30012, College Station, TX, 77842, USA
| | - Kevin Burgess
- Department of Chemistry, Texas A&M University, Box 30012, College Station, TX, 77842, USA
| |
Collapse
|
11
|
Leth JM, Leth-Espensen KZ, Kristensen KK, Kumari A, Lund Winther AM, Young SG, Ploug M. Evolution and Medical Significance of LU Domain-Containing Proteins. Int J Mol Sci 2019; 20:ijms20112760. [PMID: 31195646 PMCID: PMC6600238 DOI: 10.3390/ijms20112760] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
Proteins containing Ly6/uPAR (LU) domains exhibit very diverse biological functions and have broad taxonomic distributions in eukaryotes. In general, they adopt a characteristic three-fingered folding topology with three long loops projecting from a disulfide-rich globular core. The majority of the members of this protein domain family contain only a single LU domain, which can be secreted, glycolipid anchored, or constitute the extracellular ligand binding domain of type-I membrane proteins. Nonetheless, a few proteins contain multiple LU domains, for example, the urokinase receptor uPAR, C4.4A, and Haldisin. In the current review, we will discuss evolutionary aspects of this protein domain family with special emphasis on variations in their consensus disulfide bond patterns. Furthermore, we will present selected cases where missense mutations in LU domain-containing proteins leads to dysfunctional proteins that are causally linked to genesis of human disease.
Collapse
Affiliation(s)
- Julie Maja Leth
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Katrine Zinck Leth-Espensen
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Kristian Kølby Kristensen
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Anni Kumari
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Anne-Marie Lund Winther
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Michael Ploug
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
12
|
Leth JM, Mertens HDT, Leth-Espensen KZ, Jørgensen TJD, Ploug M. Did evolution create a flexible ligand-binding cavity in the urokinase receptor through deletion of a plesiotypic disulfide bond? J Biol Chem 2019; 294:7403-7418. [PMID: 30894413 DOI: 10.1074/jbc.ra119.007847] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/10/2019] [Indexed: 11/06/2022] Open
Abstract
The urokinase receptor (uPAR) is a founding member of a small protein family with multiple Ly6/uPAR (LU) domains. The motif defining these LU domains contains five plesiotypic disulfide bonds stabilizing its prototypical three-fingered fold having three protruding loops. Notwithstanding the detailed knowledge on structure-function relationships in uPAR, one puzzling enigma remains unexplored. Why does the first LU domain in uPAR (DI) lack one of its consensus disulfide bonds, when the absence of this particular disulfide bond impairs the correct folding of other single LU domain-containing proteins? Here, using a variety of contemporary biophysical methods, we found that reintroducing the two missing half-cystines in uPAR DI caused the spontaneous formation of the corresponding consensus 7-8 LU domain disulfide bond. Importantly, constraints due to this cross-link impaired (i) the binding of uPAR to its primary ligand urokinase and (ii) the flexible interdomain assembly of the three LU domains in uPAR. We conclude that the evolutionary deletion of this particular disulfide bond in uPAR DI may have enabled the assembly of a high-affinity urokinase-binding cavity involving all three LU domains in uPAR. Of note, an analogous neofunctionalization occurred in snake venom α-neurotoxins upon loss of another pair of the plesiotypic LU domain half-cystines. In summary, elimination of the 7-8 consensus disulfide bond in the first LU domain of uPAR did have significant functional and structural consequences.
Collapse
Affiliation(s)
- Julie M Leth
- From the Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark.,the Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Haydyn D T Mertens
- the European Molecular Biology Laboratory Hamburg, Notkestrasse 85, D-22607 Hamburg, Germany, and
| | - Katrine Zinck Leth-Espensen
- From the Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark.,the Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen N, Denmark.,the Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5320 Odense M, Denmark
| | - Thomas J D Jørgensen
- the Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5320 Odense M, Denmark
| | - Michael Ploug
- From the Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark, .,the Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
13
|
Si Y, Xu D, Bum-Erdene K, Ghozayel MK, Yang B, Clemons PA, Meroueh SO. Chemical Space Overlap with Critical Protein-Protein Interface Residues in Commercial and Specialized Small-Molecule Libraries. ChemMedChem 2019; 14:119-131. [PMID: 30548204 PMCID: PMC7175409 DOI: 10.1002/cmdc.201800537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/29/2018] [Indexed: 12/14/2022]
Abstract
There is growing interest in the use of structure-based virtual screening to identify small molecules that inhibit challenging protein-protein interactions (PPIs). In this study, we investigated how effectively chemical library members docked at the PPI interface mimic the position of critical side-chain residues known as "hot spots". Three compound collections were considered, a commercially available screening collection (ChemDiv), a collection of diversity-oriented synthesis (DOS) compounds that contains natural-product-like small molecules, and a library constructed using established reactions (the "screenable chemical universe based on intuitive data organization", SCUBIDOO). Three different tight PPIs for which hot-spot residues have been identified were selected for analysis: uPAR⋅uPA, TEAD4⋅Yap1, and CaV α⋅CaV β. Analysis of library physicochemical properties was followed by docking to the PPI receptors. A pharmacophore method was used to measure overlap between small-molecule substituents and hot-spot side chains. Fragment-like conformationally restricted small molecules showed better hot-spot overlap for interfaces with well-defined pockets such as uPAR⋅uPA, whereas better overlap was observed for more complex DOS compounds in interfaces lacking a well-defined binding site such as TEAD4⋅Yap1. Virtual screening of conformationally restricted compounds targeting uPAR⋅uPA and TEAD4⋅Yap1 followed by experimental validation reinforce these findings, as the best hits were fragment-like and had few rotatable bonds for the former, while no hits were identified for the latter. Overall, such studies provide a framework for understanding PPIs in the context of additional chemical matter and new PPI definitions.
Collapse
Affiliation(s)
- Yubing Si
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - David Xu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indianapolis, IN, 46202, USA
| | - Khuchtumur Bum-Erdene
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mona K Ghozayel
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Baocheng Yang
- Henan Provincial Key Laboratory of Nanocomposites and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, Henan, 450006, China
| | - Paul A Clemons
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
14
|
Saha N, Robev D, Mason EO, Himanen JP, Nikolov DB. Therapeutic potential of targeting the Eph/ephrin signaling complex. Int J Biochem Cell Biol 2018; 105:123-133. [PMID: 30343150 DOI: 10.1016/j.biocel.2018.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 12/27/2022]
Abstract
The Eph-ephrin signaling pathway mediates developmental processes and the proper functioning of the adult human body. This distinctive bidirectional signaling pathway includes a canonical downstream signal cascade inside the Eph-bearing cells, as well as a reverse signaling in the ephrin-bearing cells. The signaling is terminated by ADAM metalloproteinase cleavage, internalization, and degradation of the Eph/ephrin complexes. Consequently, the Eph-ephrin-ADAM signaling cascade has emerged as a key target with immense therapeutic potential particularly in the context of cancer. An interesting twist was brought forth by the emergence of ephrins as the entry receptors for the pathological Henipaviruses, which has spurred new studies to target the viral entry. The availability of high-resolution structures of the multi-modular Eph receptors in complexes with ephrins and other binding partners, such as peptides, small molecule inhibitors and antibodies, offers a wealth of information for the structure-guided development of therapeutic intervention. Furthermore, genomic data mining of Eph mutants involved in cancer provides information for targeted drug development. In this review we summarize the distinct avenues for targeting the Eph-ephrin signaling pathway, including its termination by ADAM proteinases. We highlight the latest developments in Eph-related pharmacology in the context of Eph-ephrin-ADAM-based antibodies and small molecules. Finally, the future prospects of genomics- and proteomics-based medicine are discussed.
Collapse
Affiliation(s)
- Nayanendu Saha
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Dorothea Robev
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Emilia O Mason
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Juha P Himanen
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States.
| | - Dimitar B Nikolov
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| |
Collapse
|