1
|
Awasthi A, Tiwari K, Yadav P, Bhowmick S, Tiwari DK. Synthesis of 4-styrylquinolines via direct oxidative C3-alkenylation of anthranils under Pd(II) catalysis. Chem Commun (Camb) 2024; 60:2054-2057. [PMID: 38288529 DOI: 10.1039/d3cc05790a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The palladium-catalyzed oxidative C3-alkenylation of anthranils (2,1-benzisoxazoles) with various styrenes has been successfully achieved. The C3-alkenylated anthranils were subsequently utilized in a [4+2]-cycloaddition with in situ generated α,β-unsaturated ketones leading to the synthesis of a diverse range of olefin-containing quinolines. Notably, this reaction exclusively yielded mono-alkenylated products with E-selectivity. The optimized catalytic conditions were compatible with a wide variety of substituted olefins and anthranils, forming various C3-alkenylated anthranils with good yields. To showcase the application of the present methodology, the C3-alkenylated anthranils have been employed as synthons to access a wide range of substituted quinolines.
Collapse
Affiliation(s)
- Annapurna Awasthi
- Department of Biological and Synthetic Chemistry, Center of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India. dktiwari.@cbmr.res.in
- Department of Chemistry, Institute of Science, Banaras Hindu University, 221005, Varanasi, Uttar Pradesh, India
| | - Khushboo Tiwari
- Department of Biological and Synthetic Chemistry, Center of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India. dktiwari.@cbmr.res.in
| | - Pushpendra Yadav
- Department of Biological and Synthetic Chemistry, Center of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India. dktiwari.@cbmr.res.in
- Department of Chemistry, Institute of Science, Banaras Hindu University, 221005, Varanasi, Uttar Pradesh, India
| | - Suman Bhowmick
- Department of Biological and Synthetic Chemistry, Center of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India. dktiwari.@cbmr.res.in
| | - Dharmendra Kumar Tiwari
- Department of Biological and Synthetic Chemistry, Center of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India. dktiwari.@cbmr.res.in
| |
Collapse
|
2
|
Zhou Y, Song M, Xie D, Yan S, Yu S, Xie S, Cai M, Li H, Shang L, Jiang L, Yuan C, Huang M, Li J, Xu P. Structural Dynamics-Driven Discovery of Anticancer and Antimetastatic Effects of Diltiazem and Glibenclamide Targeting Urokinase Receptor. J Med Chem 2023; 66:5415-5426. [PMID: 36854648 DOI: 10.1021/acs.jmedchem.2c01663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Diltiazem and glibenclamide are commonly used hypotensive and antidiabetic drugs. This study reports the discovery of the potential antitumor and antimetastatic effects of these two drugs using a structural dynamics-driven virtual screening targeting urokinase receptor (uPAR). Owing to uPAR's high flexibility, currently resolved crystal structures of uPAR, all in ligand-bound states, provide limited representations of its physiological conformation. To improve the accuracy of screening, we performed a long-timescale molecular dynamics simulation and obtained the representative conformations of apo-uPAR as the targets for our screening. Experimentally, we demonstrated that diltiazem and glibenclamide bound uPAR with KD values in the micromolar range. In addition, both compounds effectively suppressed tumor growth and metastasis in a uPAR-dependent manner in vitro and in vivo. This work not only provides two potent uPAR inhibitors but also reports a proof-of-concept study on the potential off-label antitumor and antimetastatic uses of diltiazem and glibenclamide.
Collapse
Affiliation(s)
- Yang Zhou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, P. R. China.,College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Meiru Song
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China.,Henan Academy of Sciences, Zhengzhou, Henan 450046, P. R. China
| | - Daoqing Xie
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Shufeng Yan
- Sanming University, Sanming, Fujian 365004, P. R. China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Song Xie
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Meiqin Cai
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Hanlin Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Le Shang
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Science, Fuzhou, Fujian 350109, P. R. China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Mingdong Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, P. R. China.,College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, P. R. China.,Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian 350108, P. R. China
| |
Collapse
|
3
|
Therapeutic Strategies Targeting Urokinase and Its Receptor in Cancer. Cancers (Basel) 2022; 14:cancers14030498. [PMID: 35158766 PMCID: PMC8833673 DOI: 10.3390/cancers14030498] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 01/19/2023] Open
Abstract
Several studies have ascertained that uPA and uPAR do participate in tumor progression and metastasis and are involved in cell adhesion, migration, invasion and survival, as well as angiogenesis. Increased levels of uPA and uPAR in tumor tissues, stroma and biological fluids correlate with adverse clinic-pathologic features and poor patient outcomes. After binding to uPAR, uPA activates plasminogen to plasmin, a broad-spectrum matrix- and fibrin-degrading enzyme able to facilitate tumor cell invasion and dissemination to distant sites. Moreover, uPAR activated by uPA regulates most cancer cell activities by interacting with a broad range of cell membrane receptors. These findings make uPA and uPAR not only promising diagnostic and prognostic markers but also attractive targets for developing anticancer therapies. In this review, we debate the uPA/uPAR structure-function relationship as well as give an update on the molecules that interfere with or inhibit uPA/uPAR functions. Additionally, the possible clinical development of these compounds is discussed.
Collapse
|
4
|
Yuan C, Guo Z, Yu S, Jiang L, Huang M. Development of inhibitors for uPAR: blocking the interaction of uPAR with its partners. Drug Discov Today 2021; 26:1076-1085. [PMID: 33486111 DOI: 10.1016/j.drudis.2021.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022]
Abstract
Urokinase-type plasminogen activator receptor (uPAR) mediates a multitude of biological activities, has key roles in several clinical indications, including malignancies and inflammation, and, thus, has attracted intensive research over the past few decades. The pleiotropic functions of uPAR can be attributed to its interaction with an array of partners. Many inhibitors have been developed to intervene with the interaction of uPAR with these partners. Here, we review the development of these classes of uPAR inhibitor and their inhibitory mechanisms to promote the translation of these inhibitors to clinical applications.
Collapse
Affiliation(s)
- Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Zhanzhi Guo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fujian, 350116, China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fujian, 350116, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fujian, 350116, China.
| |
Collapse
|
5
|
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
|
6
|
Li C, Tseng Y, Hsu T, Chang C, Hong F. Formations of aryl or pyrrole ring via palladium‐catalyzed CH functionalization on amido‐substituted quinones in the presence of amines or phosphines. J CHIN CHEM SOC-TAIP 2020. [DOI: 10.1002/jccs.202000375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Cang‐Sian Li
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| | - Yi‐Ping Tseng
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| | - Ting‐Hsuan Hsu
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| | - Chiao‐Yun Chang
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| | - Fung‐E Hong
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| |
Collapse
|
7
|
Jhou J, Cheng C, Hong F. Tertiary amines as vinyl source for the formations of aryl or pyrrole ring on
amido‐substitued
1,
4‐quinone
with the assistance of palladium salt. J CHIN CHEM SOC-TAIP 2020. [DOI: 10.1002/jccs.202000122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Jia‐Nan Jhou
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| | - Chiu‐Wen Cheng
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| | - Fung‐E Hong
- Department of Chemistry National Chung Hsing University Taichung Taiwan
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Park CK, Horton NC. Structures, functions, and mechanisms of filament forming enzymes: a renaissance of enzyme filamentation. Biophys Rev 2019; 11:927-994. [PMID: 31734826 PMCID: PMC6874960 DOI: 10.1007/s12551-019-00602-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
Filament formation by non-cytoskeletal enzymes has been known for decades, yet only relatively recently has its wide-spread role in enzyme regulation and biology come to be appreciated. This comprehensive review summarizes what is known for each enzyme confirmed to form filamentous structures in vitro, and for the many that are known only to form large self-assemblies within cells. For some enzymes, studies describing both the in vitro filamentous structures and cellular self-assembly formation are also known and described. Special attention is paid to the detailed structures of each type of enzyme filament, as well as the roles the structures play in enzyme regulation and in biology. Where it is known or hypothesized, the advantages conferred by enzyme filamentation are reviewed. Finally, the similarities, differences, and comparison to the SgrAI endonuclease system are also highlighted.
Collapse
Affiliation(s)
- Chad K. Park
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721 USA
| | - Nancy C. Horton
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721 USA
| |
Collapse
|
11
|
Small molecules inhibit ex vivo tumor growth in bone. Bioorg Med Chem 2018; 26:6128-6134. [PMID: 30470597 DOI: 10.1016/j.bmc.2018.11.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 11/10/2018] [Accepted: 11/15/2018] [Indexed: 11/22/2022]
Abstract
Bone is a common site of metastasis for breast, prostate, lung, kidney and other cancers. Bone metastases are incurable, and substantially reduce patient quality of life. To date, there exists no small-molecule therapeutic agent that can reduce tumor burden in bone. This is partly attributed to the lack of suitable in vitro assays that are good models of tumor growth in bone. Here, we take advantage of a novel ex vivo model of bone colonization to report a series of pyrrolopyrazolone small molecules that inhibit cancer cell invasion and ex vivo tumor growth in bone at single-digit micromolar concentration. We find that the compounds modulated the expression levels of genes associated with bone-forming osteoblasts, bone-destroying osteoclasts, cancer cell viability and metastasis. Our compounds provide chemical tools to uncover novel targets and pathways associated with bone metastasis, as well as for the development of compounds to prevent and reverse bone tumor growth in vivo.
Collapse
|
12
|
Wyganowska-Świątkowska M, Tarnowski M, Murtagh D, Skrzypczak-Jankun E, Jankun J. Proteolysis is the most fundamental property of malignancy and its inhibition may be used therapeutically (Review). Int J Mol Med 2018; 43:15-25. [PMID: 30431071 PMCID: PMC6257838 DOI: 10.3892/ijmm.2018.3983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 09/06/2018] [Indexed: 12/22/2022] Open
Abstract
The mortality rates of cancer patients decreased by ~1.5% per year between 2001 and 2015, although the decrease depends on patient sex, ethnic group and type of malignancy. Cancer remains a significant global health problem, requiring a search for novel treatments. The most common property of malignant tumors is their capacity to invade adjacent tissue and to metastasize, and this cancer aggressiveness is contingent on overexpression of proteolytic enzymes. The components of the plasminogen activation system (PAS) and the metal-loproteinase family [mainly matrix metalloproteinases (MMPs)] are overexpressed in malignant tumors, driving the local invasion, metastasis and angiogenesis. This is the case for numerous types of cancer, such as breast, colon, prostate and oral carcinoma, among others. Present chemotherapeutics agents typically attack all dividing cells; however, for future therapeutic agents to be clinically successful, they need to be highly selective for a specific protein(s) and act on the cancerous tissues without adverse systemic effects. Inhibition of proteolysis in cancerous tissue has the ability to attenuate tumor invasion, angiogenesis and migration. For that purpose, inhibiting both PAS and MMPs may be another approach, since the two groups of enzymes are overexpressed in cancer. In the present review, the roles and new findings on PAS and MMP families in cancer formation, growth and possible treatments are discussed.
Collapse
Affiliation(s)
| | | | - Daniel Murtagh
- Urology Research Center, Department of Urology, Health Science Campus, The University of Toledo, Toledo, OH 43614‑2598, USA
| | - Ewa Skrzypczak-Jankun
- Urology Research Center, Department of Urology, Health Science Campus, The University of Toledo, Toledo, OH 43614‑2598, USA
| | - Jerzy Jankun
- Urology Research Center, Department of Urology, Health Science Campus, The University of Toledo, Toledo, OH 43614‑2598, USA
| |
Collapse
|
13
|
Can components of the plasminogen activation system predict the outcome of kidney transplants? Cent Eur J Immunol 2018; 43:222-230. [PMID: 30135637 PMCID: PMC6102612 DOI: 10.5114/ceji.2018.77394] [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: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 12/03/2022] Open
Abstract
Proteolytic and antiproteolytic enzymes play a critical role in the physiology and pathology of different stages of human life. One of the important members of the proteolytic family is the plasminogen activation system (PAS), which includes several elements crucial for this review: the 50 kDa glycoprotein plasminogen activator inhibitor 1 (PAI-1) that inhibits tissue-type (tPA) and urokinase-type plasminogen activator (uPA). These two convert plasminogen into its active form named plasmin that can lyse a broad spectrum of proteins. Urokinase receptor (uPAR) is the binding site of uPA. This glycoprotein on the cell surface facilitates urokinase activation of plasminogen, creating high proteolytic activity close to the cell surface. PAS activities have been reported to predict the outcome of kidney transplants. However, reports on expression of PAS in kidney transplants seem to be controversial. On the one hand there are reports that impaired proteolytic activity leads to induction of chronic allograft nephropathy, while on the other hand treatment with uPA and tPA can restore function of acute renal transplants. In this comprehensive review we describe the complexity of the PAS as well as biological effects of the PAS on renal allografts, and provide a possible explanation of the reported controversy.
Collapse
|
14
|
Cai XD, Che L, Lin JX, Huang S, Li J, Liu XY, Pan XF, Wang QQ, Chen L, Lin MJ, Huang ZH, Ma HM, Wu Y, Liu SM, Zhou YB. Krüppel-like factor 17 inhibits urokinase plasminogen activator gene expression to suppress cell invasion through the Src/p38/ MAPK signaling pathway in human lung adenocarcionma. Oncotarget 2018; 8:38743-38754. [PMID: 28454121 PMCID: PMC5503568 DOI: 10.18632/oncotarget.17020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/30/2017] [Indexed: 11/25/2022] Open
Abstract
Krüppel-like factor 17 (KLF17) has been reported to be involved in invasion and metastasis suppression in lung cancer, but the molecular mechanisms underlying the anti-invasion and anti-metastasis roles of KLF17 in lung cancer are not fully illustrated. Here, we showed that KLF17 inhibited the invasion of A549 and H322 cells; the anti-invasion effect of KLF17 was associated with the suppression of urokinase plasminogen activator (uPA/PLAU) expression. KLF17 can bind with the promoter of uPA and inhibit its expression. Enforced expression of uPA abrogated the anti-invasion effect of KLF17 in A549 and H322 cells. In addition, immunohistochemistry staining showed that the protein expression of KLF17 was negatively correlated with that of uPA in archived samples from patients with lymph node metastasis of lung adenocarcinoma (rho = −0.62, P = 0.01). The mutually exclusive expression of KLF17 with uPA could predict lymph node metastasis for lung adenocarcinoma (AUC = 0.758, P = 0.005). Enforced expression of KLF17 inhibited the expression of phosphorylated Src and phosphorylated p38/MAPK in A549 and H322 cells. The invasiveness of the cells were suppressed by treating with sb203580 (p38/MAPK inhibitor) or HY-13805 (PP2, Src inhibitor). furthermore, p38/MAPK inhibition could block the KLF17-induced reduction of p-p38/MAPK and uPA, and Src inhibition enhanced the KLF17-induced suppression of p-Src and uPA in A549 and H322 cells. In conclusion, our study indicated that KLF17 suppressed the uPA-mediated invasion of lung adenocarcinoma. The Src and p38/MAPK signaling pathways were suggested as mediators of KLF17-induced uPA inhibition, thus providing evidence that KLF17 might be a potential anti-invasion candidate for lung adenocarcinoma.
Collapse
Affiliation(s)
- Xing-Dong Cai
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Li Che
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jia-Xin Lin
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuai Huang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jiong Li
- Department of Anatomy, The Medical College of Jinan University, Guangzhou 510630, China
| | - Xiao-Yan Liu
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xing-Fei Pan
- Department of Infectious Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Qin-Qin Wang
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Li Chen
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ming-Juan Lin
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhi-Hong Huang
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hong-Ming Ma
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yi Wu
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Sheng-Ming Liu
- Department of Respiratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yan-Bin Zhou
- Department of Pulmonary Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
15
|
Sk MR, Chakraborty S, Mal D. Studies directed toward total synthesis of rhodocomatulins: A regioselective synthesis of brominated hydroxyanthraquinones by anionic annulations. SYNTHETIC COMMUN 2018. [DOI: 10.1080/00397911.2017.1401637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Md Raja Sk
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Soumen Chakraborty
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Dipakranjan Mal
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
16
|
Xu D, Si Y, Meroueh SO. A Computational Investigation of Small-Molecule Engagement of Hot Spots at Protein-Protein Interaction Interfaces. J Chem Inf Model 2017; 57:2250-2272. [PMID: 28766941 DOI: 10.1021/acs.jcim.7b00181] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The binding affinity of a protein-protein interaction is concentrated at amino acids known as hot spots. It has been suggested that small molecules disrupt protein-protein interactions by either (i) engaging receptor protein hot spots or (ii) mimicking hot spots of the protein ligand. Yet, no systematic studies have been done to explore how effectively existing small-molecule protein-protein interaction inhibitors mimic or engage hot spots at protein interfaces. Here, we employ explicit-solvent molecular dynamics simulations and end-point MM-GBSA free energy calculations to explore this question. We select 36 compounds for which high-quality binding affinity and cocrystal structures are available. Five complexes that belong to three classes of protein-protein interactions (primary, secondary, and tertiary) were considered, namely, BRD4•H4, XIAP•Smac, MDM2•p53, Bcl-xL•Bak, and IL-2•IL-2Rα. Computational alanine scanning using MM-GBSA identified hot-spot residues at the interface of these protein interactions. Decomposition energies compared the interaction of small molecules with individual receptor hot spots to those of the native protein ligand. Pharmacophore analysis was used to investigate how effectively small molecules mimic the position of hot spots of the protein ligand. Finally, we study whether small molecules mimic the effects of the native protein ligand on the receptor dynamics. Our results show that, in general, existing small-molecule inhibitors of protein-protein interactions do not optimally mimic protein-ligand hot spots, nor do they effectively engage protein receptor hot spots. The more effective use of hot spots in future drug design efforts may result in smaller compounds with higher ligand efficiencies that may lead to greater success in clinical trials.
Collapse
Affiliation(s)
- David Xu
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing , Indianapolis, Indiana 46202, United States
| | | | | |
Collapse
|
17
|
Patil VV, Gayakwad EM, Patel KP, Shankarling GS. Efficient, facile metal free protocols for the bromination of commercially important deactivated aminoanthracene-9,10-diones. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.05.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
18
|
Liu D, Xu D, Liu M, Knabe WE, Yuan C, Zhou D, Huang M, Meroueh SO. Small Molecules Engage Hot Spots through Cooperative Binding To Inhibit a Tight Protein-Protein Interaction. Biochemistry 2017; 56:1768-1784. [PMID: 28186725 DOI: 10.1021/acs.biochem.6b01039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein-protein interactions drive every aspect of cell signaling, yet only a few small-molecule inhibitors of these interactions exist. Despite our ability to identify critical residues known as hot spots, little is known about how to effectively engage them to disrupt protein-protein interactions. Here, we take advantage of the ease of preparation and stability of pyrrolinone 1, a small-molecule inhibitor of the tight interaction between the urokinase receptor (uPAR) and its binding partner, the urokinase-type plasminogen activator uPA, to synthesize more than 40 derivatives and explore their effect on the protein-protein interaction. We report the crystal structure of uPAR bound to previously discovered pyrazole 3 and to pyrrolinone 12. While both 3 and 12 bind to uPAR and compete with a fluorescently labeled peptide probe, only 12 and its derivatives inhibit the full uPAR·uPA interaction. Compounds 3 and 12 mimic and engage different hot-spot residues on uPA and uPAR, respectively. Interestingly, 12 is involved in a π-cation interaction with Arg-53, which is not considered a hot spot. Explicit-solvent molecular dynamics simulations reveal that 3 and 12 exhibit dramatically different correlations of motion with residues on uPAR. Free energy calculations for the wild-type and mutant uPAR bound to uPA or 12 show that Arg-53 interacts with uPA or with 12 in a highly cooperative manner, thereby altering the contributions of hot spots to uPAR binding. The direct engagement of peripheral residues not considered hot spots through π-cation or salt-bridge interactions could provide new opportunities for enhanced small-molecule engagement of hot spots to disrupt challenging protein-protein interactions.
Collapse
Affiliation(s)
- Degang Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - David Xu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States.,Department of BioHealth Informatics, Indiana University School of Informatics and Computing , Indianapolis, Indiana 46202, United States
| | - Min Liu
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Science , Gulou District, Fuzhou, Fujian 3500002, China
| | - William Eric Knabe
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Cai Yuan
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Science , Gulou District, Fuzhou, Fujian 3500002, China
| | - Donghui Zhou
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Mingdong Huang
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Science , Gulou District, Fuzhou, Fujian 3500002, China
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| |
Collapse
|
19
|
Montuori N, Pesapane A, Rossi FW, Giudice V, De Paulis A, Selleri C, Ragno P. Urokinase type plasminogen activator receptor (uPAR) as a new therapeutic target in cancer. Transl Med UniSa 2016; 15:15-21. [PMID: 27896223 PMCID: PMC5120746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The urokinase (uPA)-type plasminogen activator receptor (uPAR) is a GPI-anchored receptor that focuses urokinase (uPA) proteolytic activity on the cell surface. uPAR also regulates cell adhesion, migration and proliferation, protects from apoptosis and contributes to epithelial mesenchymal transition (EMT), independently of uPA enzymatic activity. Indeed, uPAR interacts with beta1, beta2 and beta3 integrins, thus regulating their activities. uPAR cross-talks with receptor tyrosine kinases through integrins and regulates cancer cell dormancy, proliferation and angiogenesis. Moreover, uPAR mediates uPA-dependent cell migration and chemotaxis induced by fMet-Leu-Phe (fMLF), through its association with fMLF-receptors (fMLF-Rs). Further, uPAR is an adhesion receptor because it binds vitronectin (VN), a component of provisional extracellular matrix. High uPAR expression predicts for more aggressive disease in several cancer types for its ability to increase invasion and metastasis. In fact, uPAR has been hypothesized to be the link between tumor cell dormancy and proliferation that usually precedes the onset of metastasis. Thus, inhibiting uPAR could be a feasible approach to affect tumor growth and metastasis. Here, we review the more recent advances in the development of uPAR-targeted anti-cancer therapeutic agents suitable for further optimization or ready for the evaluation in early clinical trials.
Collapse
Affiliation(s)
- Nunzia Montuori
- Department of Translational Medical Sciences, University Federico II, Naples, Italy
| | - Ada Pesapane
- Department of Translational Medical Sciences, University Federico II, Naples, Italy
| | - Francesca W Rossi
- Department of Translational Medical Sciences, University Federico II, Naples, Italy
| | - Valentina Giudice
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Amato De Paulis
- Department of Translational Medical Sciences, University Federico II, Naples, Italy
| | - Carmine Selleri
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, Salerno, Italy
| |
Collapse
|
20
|
Rullo AF, Fitzgerald KJ, Muthusamy V, Liu M, Yuan C, Huang M, Kim M, Cho AE, Spiegel DA. Re-engineering the Immune Response to Metastatic Cancer: Antibody-Recruiting Small Molecules Targeting the Urokinase Receptor. Angew Chem Int Ed Engl 2016; 55:3642-6. [PMID: 26879524 PMCID: PMC5597302 DOI: 10.1002/anie.201510866] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/23/2015] [Indexed: 12/19/2022]
Abstract
Developing selective strategies to treat metastatic cancers remains a significant challenge. Herein, we report the first antibody-recruiting small molecule (ARM) that is capable of recognizing the urokinase-type plasminogen activator receptor (uPAR), a uniquely overexpressed cancer cell-surface marker, and facilitating the immune-mediated destruction of cancer cells. A co-crystal structure of the ARM-U2/uPAR complex was obtained, representing the first crystal structure of uPAR complexed with a non-peptide ligand. Finally, we demonstrated that ARM-U2 substantially suppresses tumor growth in vivo with no evidence of weight loss, unlike the standard-of-care agent doxorubicin. This work underscores the promise of antibody-recruiting molecules as immunotherapeutics for treating cancer.
Collapse
Affiliation(s)
- Anthony F Rullo
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, CT, 06511, USA
| | - Kelly J Fitzgerald
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Viswanathan Muthusamy
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, CT, 06511, USA
| | - Min Liu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, 155 Yang Qiao West Road, Fuzhou, Fujian, 350002, China
| | - Cai Yuan
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, 155 Yang Qiao West Road, Fuzhou, Fujian, 350002, China
| | - Mingdong Huang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, 155 Yang Qiao West Road, Fuzhou, Fujian, 350002, China
| | - Minsup Kim
- Department of Bioinformatics, Korea University, 2511 Sejong-ro, Sejong, 339-700, Korea
| | - Art E Cho
- Department of Bioinformatics, Korea University, 2511 Sejong-ro, Sejong, 339-700, Korea
| | - David A Spiegel
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, CT, 06511, USA.
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
21
|
Rullo AF, Fitzgerald KJ, Muthusamy V, Liu M, Yuan C, Huang M, Kim M, Cho AE, Spiegel DA. Re-engineering the Immune Response to Metastatic Cancer: Antibody-Recruiting Small Molecules Targeting the Urokinase Receptor. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201510866] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Anthony F. Rullo
- Department of Chemistry; Yale University; 225 Prospect Street New Haven CT 06511 USA
| | - Kelly J. Fitzgerald
- Department of Pharmacology; Yale School of Medicine; 333 Cedar Street New Haven CT 06520 USA
| | - Viswanathan Muthusamy
- Department of Chemistry; Yale University; 225 Prospect Street New Haven CT 06511 USA
| | - Min Liu
- State Key Laboratory of Structural Chemistry; Fujian Institute of Research on the Structure of Matter; Chinese Academy of Sciences; 155 Yang Qiao West Road Fuzhou Fujian 350002 China
| | - Cai Yuan
- State Key Laboratory of Structural Chemistry; Fujian Institute of Research on the Structure of Matter; Chinese Academy of Sciences; 155 Yang Qiao West Road Fuzhou Fujian 350002 China
| | - Mingdong Huang
- State Key Laboratory of Structural Chemistry; Fujian Institute of Research on the Structure of Matter; Chinese Academy of Sciences; 155 Yang Qiao West Road Fuzhou Fujian 350002 China
| | - Minsup Kim
- Department of Bioinformatics; Korea University; 2511 Sejong-ro Sejong 339-700 Korea
| | - Art E. Cho
- Department of Bioinformatics; Korea University; 2511 Sejong-ro Sejong 339-700 Korea
| | - David A. Spiegel
- Department of Chemistry; Yale University; 225 Prospect Street New Haven CT 06511 USA
- Department of Pharmacology; Yale School of Medicine; 333 Cedar Street New Haven CT 06520 USA
| |
Collapse
|
22
|
Chen Y, Gong L, Gao N, Liao J, Sun J, Wang Y, Wang L, Zhu P, Fan Q, Wang YA, Zeng W, Mao H, Yang L, Gao F. Preclinical evaluation of a urokinase plasminogen activator receptor-targeted nanoprobe in rhesus monkeys. Int J Nanomedicine 2015; 10:6689-98. [PMID: 26604745 PMCID: PMC4630189 DOI: 10.2147/ijn.s90587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose To translate a recombinant peptide containing the amino-terminal fragment (ATF) of urokinase plasminogen activator receptor-targeted magnetic iron oxide (IO) nanoparticles (uPAR-targeted human ATF-IONPs) into clinical applications, we conducted a pilot study to evaluate the toxicity and pharmacokinetics of this nanoparticle in normal rhesus monkeys. Methods We assessed the changes in the following: magnetic resonance imaging (MRI) signals from pretreatment stage to 14 days posttreatment, serum iron concentrations from 5 minutes posttreatment to 12 weeks posttreatment, routine blood examination and serum chemistry analysis results from pretreatment stage to 12 weeks after administration, and results of staining of the liver with Perls’ Prussian Blue and hematoxylin–eosin at 24 hours and 3 months posttreatment in two rhesus monkeys following an intravenous administration of the targeted nanoparticles either with a polyethylene glycol (ATF-PEG-IONP) or without a PEG (ATF-IONP) coating. Results The levels of alkaline phosphatase, alanine transaminase, and direct bilirubin in the two monkeys increased immediately after the administration of the IONPs but returned to normal within 20 days and stayed within the normal reference range 3 months after the injection. The creatinine levels of the two monkeys stayed within the normal range during the study. In addition, red blood cells, white blood cells, hemoglobin level, and platelets remained normal during the 3 months of the study. Conclusion All of the results suggest that a transient injury in terms of normal organ functions, but no microscopic necrotic lesions, was observed at a systemic delivery dose of 5 mg/kg of iron equivalent concentration in the acute phase, and that no chronic toxicity was found 3 months after the injection. Therefore, we conclude that uPAR-targeted IONPs have the potential to be used as receptor-targeted MRI contrasts as well as theranostic agents for the detection and treatment of human cancers in future studies.
Collapse
Affiliation(s)
- Yushu Chen
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Li Gong
- Sichuan Primed Bio-Tech Group Co, Ltd, Chengdu, People's Republic of China
| | - Ning Gao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Jichun Liao
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jiayu Sun
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yuqing Wang
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lei Wang
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Pengjin Zhu
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qing Fan
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | | | - Wen Zeng
- Sichuan Primed Bio-Tech Group Co, Ltd, Chengdu, People's Republic of China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Fabao Gao
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
23
|
Liu D, Zhou D, Wang B, Knabe WE, Meroueh SO. A new class of orthosteric uPAR·uPA small-molecule antagonists are allosteric inhibitors of the uPAR·vitronectin interaction. ACS Chem Biol 2015; 10:1521-34. [PMID: 25671694 DOI: 10.1021/cb500832q] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The urokinase receptor (uPAR) is a GPI-anchored cell surface receptor that is at the center of an intricate network of protein-protein interactions. Its immediate binding partners are the serine proteinase urokinase (uPA), and vitronectin (VTN), a component of the extracellular matrix. uPA and VTN bind at distinct sites on uPAR to promote extracellular matrix degradation and integrin signaling, respectively. Here, we report the discovery of a new class of pyrrolone small-molecule inhibitors of the tight ∼1 nM uPAR·uPA protein-protein interaction. These compounds were designed to bind to the uPA pocket on uPAR. The highest affinity compound, namely 7, displaced a fluorescently labeled α-helical peptide (AE147-FAM) with an inhibition constant Ki of 0.7 μM and inhibited the tight uPAR·uPAATF interaction with an IC50 of 18 μM. Biophysical studies with surface plasmon resonance showed that VTN binding is highly dependent on uPA. This cooperative binding was confirmed as 7, which binds at the uPAR·uPA interface, also inhibited the distal VTN·uPAR interaction. In cell culture, 7 blocked the uPAR·uPA interaction in uPAR-expressing human embryonic kidney (HEK-293) cells and impaired cell adhesion to VTN, a process that is mediated by integrins. As a result, 7 inhibited integrin signaling in MDA-MB-231 cancer cells as evidenced by a decrease in focal adhesion kinase (FAK) phosphorylation and Rac1 GTPase activation. Consistent with these results, 7 blocked breast MDA-MB-231 cancer cell invasion with IC50 values similar to those observed in ELISA and surface plasmon resonance competition studies. Explicit-solvent molecular dynamics simulations show that the cooperativity between uPA and VTN is attributed to stabilization of uPAR motion by uPA. In addition, free energy calculations revealed that uPA stabilizes the VTNSMB·uPAR interaction through more favorable electrostatics and entropy. Disruption of the uPAR·VTNSMB interaction by 7 is consistent with the cooperative binding to uPAR by uPA and VTN. Interestingly, the VTNSMB·uPAR interaction was less favorable in the VTNSMB·uPAR·7 complex suggesting potential cooperativity between 7 and VTN. Compound 7 provides an excellent starting point for the development of more potent derivatives to explore uPAR biology.
Collapse
Affiliation(s)
| | | | - Bo Wang
- Department
of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis (IUPUI), Indianapolis, Indiana 46202, United States
| | | | - Samy O. Meroueh
- Department
of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis (IUPUI), Indianapolis, Indiana 46202, United States
| |
Collapse
|
24
|
|
25
|
McMahon BJ, Kwaan HC. Components of the Plasminogen-Plasmin System as Biologic Markers for Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 867:145-56. [PMID: 26530365 DOI: 10.1007/978-94-017-7215-0_10] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Members of the plasminogen-plasmin (PP) system participate in many physiologic functions. In particular, uPA, its receptor (uPAR) and its inhibitor PAI-1 play an important role in cell migration, cell proliferation and tissue remodeling. Through a number of interactions, these components of the PP system are also involved in the pathogenesis of many diseases. In cancer, they modulate the essential processes of tumor development, growth, invasion and metastasis as well as angiogenesis and fibrosis. Thus, quantification of uPA, uPAR and PAI-1 in tumors and, in some cases in the circulating blood, became of potential value in the prognostication of many types of cancer. These include cancer of the breast, stomach, colon and rectum, esophagus, pancreas, glioma, lung, kidney, prostate, uterine cervix, ovary, liver and bone. Published data are reviewed in this chapter. Clinical validation of the prognostic value has also been made, particularly in cancer of the breast. Inclusion of these biomarkers in the risk assessment of cancer patients is now considered in the risk-adapted management in carcinoma of the breast. Factors limiting its broader use are discussed with suggestions how these can be overcome. Hopefully the use of these biomarkers will be applied to other types of cancer in the near future.
Collapse
Affiliation(s)
- Brandon J McMahon
- Division of Hematology/Oncology, Feinberg School of Medicine, and the Robert H. Lurie Cancer, Northwestern University, Chicago, IL, USA.,Olson Pavilion, Room 8258, 710 N. Fairbanks Court, Chicago, IL, 60611, USA
| | - Hau C Kwaan
- Division of Hematology/Oncology, Feinberg School of Medicine, and the Robert H. Lurie Cancer, Northwestern University, Chicago, IL, USA. .,Olson Pavilion, Room 8258, 710 N. Fairbanks Court, Chicago, IL, 60611, USA.
| |
Collapse
|
26
|
Xiang Y, Li Q, Huang D, Tang X, Wang L, Shi Y, Zhang W, Yang T, Xiao C, Wang J. Preparation and antitumor effect of a toxin-linked conjugate targeting vascular endothelial growth factor receptor and urokinase plasminogen activator. Exp Biol Med (Maywood) 2014; 240:160-8. [PMID: 25125500 DOI: 10.1177/1535370214547154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The aberrant signaling activation of vascular endothelial growth factor receptor (VEGFR) and urokinase plasminogen activator (uPA) is a common characteristic of many tumors, including lung cancer. Accordingly, VEGFR and uPA have emerged as attractive targets for tumor. KDR (Flk-1/VEGFR-2), a member of the VEGFR family, has been recognized as an important target for antiangiogenesis in tumor. In this study, a recombinant immunotoxin was produced to specifically target KDR-expressing tumor vascular endothelial cells and uPA-expressing tumor cells and mediate antitumor angiogenesis and antitumor effect. Based on its potent inhibitory effect on protein synthesis, Luffin-beta (Lβ) ribosome-inactivating protein was selected as part of a recombinant fusion protein, a single-chain variable fragment against KDR (KDRscFv)-uPA cleavage site (uPAcs)-Lβ-KDEL (named as KPLK). The KDRscFv-uPAcs-Lβ-KDEL (KPLK) contained a single-chain variable fragment (scFv) against KDR, uPAcs, Lβ, and the retention signal for endoplasmic reticulum proteins KDEL (Lys-Asp-Glu-Leu). The KPLK-expressing vector was expressed in Escherichia coli, and the KPLK protein was isolated with nickel affinity chromatography and gel filtration chromatography. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis test demonstrated KPLK was effectively expressed. Result of in vitro cell viability assay on non-small cell lung cancer (NSCLC) H460 cell line (uPA-positive cell) revealed that KPLK significantly inhibited cell proliferation, induced apoptosis, and accumulated cells in S and G2/M phases, but the normal cell line (human submandibular gland cell) was unaffected. These effects were enhanced when uPA was added to digest KPLK to release Lβ. For in vivo assay of KPLK, subcutaneous xenograft tumor model of nude mice were established with H460 cells. Growth of solid tumors was significantly inhibited in animals treated with KPLK up to 21 days, tumor weights were decreased, and the expression of angiogenesis marker CD31 was downregulated; meanwhile, the apoptosis-related protein casspase-3 was upregulated. These results suggested that the recombinant KPLK may have therapeutic applications on tumors, especially uPA-overexpressing ones.
Collapse
Affiliation(s)
- Ying Xiang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Qiying Li
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Dehong Huang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Xianjun Tang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Li Wang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Yang Shi
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Wenjun Zhang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Tao Yang
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Chunyan Xiao
- Department of Biotherapy and Hemo-oncology, Chongqing Cancer Institute, Chongqing 400030, China
| | - Jianghong Wang
- Center of Endoscopy Examination & Therapy, Chongqing Cancer Institute, Chongqing 400030, China
| |
Collapse
|
27
|
Patil VV, Shankarling GS. Nonanebis(peroxoic acid): a stable peracid for oxidative bromination of aminoanthracene-9,10-dione. Beilstein J Org Chem 2014; 10:921-8. [PMID: 24991241 PMCID: PMC4077418 DOI: 10.3762/bjoc.10.90] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/01/2014] [Indexed: 12/01/2022] Open
Abstract
A new protocol for the oxidative bromination of aminoanthracene-9,10-dione, which is highly deactivated towards the electrophilic substitution is investigated. The peracid, nonanebis(peroxoic acid), possesses advantages such as better stability at room temperature, it is easy to prepare and non-shock sensitiv as compared to the conventional peracids. The present protocol has a broad scope for the bromination of various substituted and unsubstituted aminoanthracene-9,10-diones.
Collapse
Affiliation(s)
- Vilas Venunath Patil
- Department of Dyestuff Technology, Institute of Chemical Technology, N. P. Marg, Matunga, Mumbai - 400019, India. Tel.: 91-22-33612708
| | - Ganapati Subray Shankarling
- Department of Dyestuff Technology, Institute of Chemical Technology, N. P. Marg, Matunga, Mumbai - 400019, India. Tel.: 91-22-33612708
| |
Collapse
|
28
|
Mani T, Liu D, Zhou D, Li L, Knabe WE, Wang F, Oh K, Meroueh SO. Probing binding and cellular activity of pyrrolidinone and piperidinone small molecules targeting the urokinase receptor. ChemMedChem 2013; 8:1963-77. [PMID: 24115356 DOI: 10.1002/cmdc.201300340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Indexed: 01/16/2023]
Abstract
The urokinase receptor (uPAR) is a cell-surface protein that is part of an intricate web of transient and tight protein interactions that promote cancer cell invasion and metastasis. Here, we evaluate the binding and biological activity of a new class of pyrrolidinone and piperidinone compounds, along with derivatives of previously-identified pyrazole and propylamine compounds. Competition assays revealed that the compounds displace a fluorescently labeled peptide (AE147-FAM) with inhibition constant (Ki ) values ranging from 6 to 63 μM. Structure-based computational pharmacophore analysis followed by extensive explicit-solvent molecular dynamics (MD) simulations and free energy calculations suggested the pyrazole-based and piperidinone-based compounds adopt different binding modes, despite their similar two-dimensional structures. In cells, pyrazole-based compounds showed significant inhibition of breast adenocarcinoma (MDA-MB-231) and pancreatic ductal adenocarcinoma (PDAC) cell proliferation, but piperidinone-containing compounds exhibited no cytotoxicity even at concentrations of 100 μM. One pyrazole-based compound impaired MDA-MB-231 invasion, adhesion, and migration in a concentration-dependent manner, while the piperidinone inhibited only invasion. The pyrazole derivative inhibited matrix metalloprotease-9 (gelatinase) activity in a concentration-dependent manner, while the piperidinone showed no effect suggesting different mechanisms for inhibition of cell invasion. Signaling studies further highlighted these differences, showing that pyrazole compounds completely inhibited ERK phosphorylation and impaired HIF1α and NF-κB signaling, while pyrrolidinones and piperidinones had no effect. Annexin V staining suggested that the effect of the pyrazole-based compound on proliferation was due to cell killing through an apoptotic mechanism. The compounds identified represent valuable leads in the design of further derivatives with higher affinities and potential probes to unravel the protein-protein interactions of uPAR.
Collapse
Affiliation(s)
- Timmy Mani
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 (USA)
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Rea VEA, Lavecchia A, Di Giovanni C, Rossi FW, Gorrasi A, Pesapane A, de Paulis A, Ragno P, Montuori N. Discovery of new small molecules targeting the vitronectin-binding site of the urokinase receptor that block cancer cell invasion. Mol Cancer Ther 2013; 12:1402-16. [PMID: 23699658 DOI: 10.1158/1535-7163.mct-12-1249] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Besides focusing urokinase (uPA) proteolytic activity on the cell membrane, the uPA receptor (uPAR) is able to bind vitronectin, via a direct binding site. Furthermore, uPAR interacts with other cell surface receptors, such as integrins, receptor tyrosine kinases, and chemotaxis receptors, triggering cell-signaling pathways that promote tumor progression. The ability of uPAR to coordinate binding and degradation of extracellular matrix (ECM) and cell signaling makes it an attractive therapeutic target in cancer. We used structure-based virtual screening (SB-VS) to search for small molecules targeting the uPAR-binding site for vitronectin. Forty-one compounds were identified and tested on uPAR-negative HEK-293 epithelial cells transfected with uPAR (uPAR-293 cells), using the parental cell line transfected with the empty vector (V-293 cells) as a control. Compounds 6 and 37 selectively inhibited uPAR-293 cell adhesion to vitronectin and the resulting changes in cell morphology and signal transduction, without exerting any effect on V-293 cells. Compounds 6 and 37 inhibited uPAR-293 cell binding to vitronectin with IC50 values of 3.6 and 1.2 μmol/L, respectively. Compounds 6 and 37 targeted S88 and R91, key residues for uPAR binding to vitronectin but also for uPAR interaction with the fMLF family of chemotaxis receptors (fMLF-Rs). As a consequence, compounds 6 and 37 impaired uPAR-293 cell migration toward fetal calf serum (FCS), uPA, and fMLF, likely by inhibiting the interaction between uPAR and FPR1, the high affinity fMLF-R. Both compounds blocked in vitro ECM invasion of several cancer cell types, thus representing new promising leads for pharmaceuticals in cancer.
Collapse
|
30
|
Mani T, Wang F, Knabe WE, Sinn AL, Khanna M, Jo I, Sandusky GE, Sledge GW, Jones DR, Khanna R, Pollok KE, Meroueh SO. Small-molecule inhibition of the uPAR·uPA interaction: synthesis, biochemical, cellular, in vivo pharmacokinetics and efficacy studies in breast cancer metastasis. Bioorg Med Chem 2013; 21:2145-55. [PMID: 23411397 DOI: 10.1016/j.bmc.2012.12.047] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/13/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
The uPAR·uPA protein-protein interaction (PPI) is involved in signaling and proteolytic events that promote tumor invasion and metastasis. A previous study had identified 4 (IPR-803) from computational screening of a commercial chemical library and shown that the compound inhibited uPAR·uPA PPI in competition biochemical assays and invasion cellular studies. Here, we synthesize 4 to evaluate in vivo pharmacokinetic (PK) and efficacy studies in a murine breast cancer metastasis model. First, we show, using fluorescence polarization and saturation transfer difference (STD) NMR, that 4 binds directly to uPAR with sub-micromolar affinity of 0.2 μM. We show that 4 blocks invasion of breast MDA-MB-231, and inhibits matrix metalloproteinase (MMP) breakdown of the extracellular matrix (ECM). Derivatives of 4 also inhibited MMP activity and blocked invasion in a concentration-dependent manner. Compound 4 also impaired MDA-MB-231 cell adhesion and migration. Extensive in vivo PK studies in NOD-SCID mice revealed a half-life of nearly 5h and peak concentration of 5 μM. Similar levels of the inhibitor were detected in tumor tissue up to 10h. Female NSG mice inoculated with highly malignant TMD-MDA-MB-231 in their mammary fat pads showed that 4 impaired metastasis to the lungs with only four of the treated mice showing severe or marked metastasis compared to ten for the untreated mice. Compound 4 is a promising template for the development of compounds with enhanced PK parameters and greater efficacy.
Collapse
Affiliation(s)
- Timmy Mani
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Van Nuys Medical Science Building, MS 4023, 635 Barnhill Drive, Indianapolis, IN 46202-5122, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|