1
|
Jaiswal AK, Thaxton ML, Scherer GM, Sorrentino JP, Garg NK, Rao DS. Small molecule inhibition of RNA binding proteins in haematologic cancer. RNA Biol 2024; 21:1-14. [PMID: 38329136 PMCID: PMC10857685 DOI: 10.1080/15476286.2024.2303558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/18/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024] Open
Abstract
In recent years, advances in biomedicine have revealed an important role for post-transcriptional mechanisms of gene expression regulation in pathologic conditions. In cancer in general and leukaemia specifically, RNA binding proteins have emerged as important regulator of RNA homoeostasis that are often dysregulated in the disease state. Having established the importance of these pathogenetic mechanisms, there have been a number of efforts to target RNA binding proteins using oligonucleotide-based strategies, as well as with small organic molecules. The field is at an exciting inflection point with the convergence of biomedical knowledge, small molecule screening strategies and improved chemical methods for synthesis and construction of sophisticated small molecules. Here, we review the mechanisms of post-transcriptional gene regulation, specifically in leukaemia, current small-molecule based efforts to target RNA binding proteins, and future prospects.
Collapse
Affiliation(s)
- Amit K. Jaiswal
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Michelle L. Thaxton
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Georgia M. Scherer
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Jacob P. Sorrentino
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Neil K. Garg
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Dinesh S. Rao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Wu X, Zhang Q, Guo Y, Zhang H, Guo X, You Q, Wang L. Methods for the Discovery and Identification of Small Molecules Targeting Oxidative Stress-Related Protein–Protein Interactions: An Update. Antioxidants (Basel) 2022; 11:antiox11040619. [PMID: 35453304 PMCID: PMC9025695 DOI: 10.3390/antiox11040619] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
The oxidative stress response pathway is one of the hotspots of current pharmaceutical research. Many proteins involved in these pathways work through protein–protein interactions (PPIs). Hence, targeting PPI to develop drugs for an oxidative stress response is a promising strategy. In recent years, small molecules targeting protein–protein interactions (PPIs), which provide efficient methods for drug discovery, are being investigated by an increasing number of studies. However, unlike the enzyme–ligand binding mode, PPIs usually exhibit large and dynamic binding interfaces, which raise additional challenges for the discovery and optimization of small molecules and for the biochemical techniques used to screen compounds and study structure–activity relationships (SARs). Currently, multiple types of PPIs have been clustered into different classes, which make it difficult to design stationary methods for small molecules. Deficient experimental methods are plaguing medicinal chemists and are becoming a major challenge in the discovery of PPI inhibitors. In this review, we present current methods that are specifically used in the discovery and identification of small molecules that target oxidative stress-related PPIs, including proximity-based, affinity-based, competition-based, structure-guided, and function-based methods. Our aim is to introduce feasible methods and their characteristics that are implemented in the discovery of small molecules for different types of PPIs. For each of these methods, we highlight successful examples of PPI inhibitors associated with oxidative stress to illustrate the strategies and provide insights for further design.
Collapse
Affiliation(s)
- Xuexuan Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuqi Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hengheng Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| |
Collapse
|
3
|
Cryan LM, Tsang TM, Stiles J, Bazinet L, Lee SL, Garrard S, Madrian E, Roberts C, Payne J, Jensen A, Frankel AE, Ackroyd PC, Christensen KA, Rogers MS. Capillary morphogenesis gene 2 (CMG2) mediates growth factor-induced angiogenesis by regulating endothelial cell chemotaxis. Angiogenesis 2022; 25:397-410. [PMID: 35212873 PMCID: PMC9250616 DOI: 10.1007/s10456-022-09833-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/06/2022] [Indexed: 11/28/2022]
Abstract
Anthrax protective antigen (PA) is a potent inhibitor of pathological angiogenesis with an unknown mechanism. In anthrax intoxication, PA interacts with capillary morphogenesis gene 2 (CMG2) and tumor endothelial marker 8 (TEM8). Here, we show that CMG2 mediates the antiangiogenic effects of PA and is required for growth-factor-induced chemotaxis. Using specific inhibitors of CMG2 and TEM8 interaction with natural ligand, as well as mice with the CMG2 or TEM8 transmembrane and intracellular domains disrupted, we demonstrate that inhibiting CMG2, but not TEM8 reduces growth-factor-induced angiogenesis in the cornea. Furthermore, the antiangiogenic effect of PA was abolished when the CMG2, but not the TEM8, gene was disrupted. Binding experiments demonstrated a broad ligand specificity for CMG2 among extracellular matrix (ECM) proteins. Ex vivo experiments demonstrated that CMG2 (but not TEM8) is required for PA activity in human dermal microvascular endothelial cell (HMVEC-d) network formation assays. Remarkably, blocking CMG2-ligand binding with PA or CRISPR knockout abolishes endothelial cell chemotaxis but not chemokinesis in microfluidic migration assays. These effects are phenocopied by Rho inhibition. Because CMG2 mediates the chemotactic response of endothelial cells to peptide growth factors in an ECM-dependent fashion, CMG2 is well-placed to integrate growth factor and ECM signals. Thus, CMG2 targeting is a novel way to inhibit angiogenesis.
Collapse
Affiliation(s)
- Lorna M Cryan
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, 11.211 Karp Family Research Bldg., 300 Longwood Ave., Boston, MA, 02115, USA
| | - Tsz-Ming Tsang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Jessica Stiles
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, 11.211 Karp Family Research Bldg., 300 Longwood Ave., Boston, MA, 02115, USA
| | - Lauren Bazinet
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, 11.211 Karp Family Research Bldg., 300 Longwood Ave., Boston, MA, 02115, USA
| | - Sai Lun Lee
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Samuel Garrard
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA.,Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, 11.211 Karp Family Research Bldg., 300 Longwood Ave., Boston, MA, 02115, USA
| | - Erika Madrian
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, 11.211 Karp Family Research Bldg., 300 Longwood Ave., Boston, MA, 02115, USA
| | - Cody Roberts
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Jessie Payne
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Andrew Jensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Arthur E Frankel
- Department of Medicine, West Palm Beach VA Medical Center, 7305 N Military Trail, West Palm Beach, FL, 33410, USA
| | - P Christine Ackroyd
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Kenneth A Christensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Michael S Rogers
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, 11.211 Karp Family Research Bldg., 300 Longwood Ave., Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Liu L, He F, Yu Y, Wang Y. Application of FRET Biosensors in Mechanobiology and Mechanopharmacological Screening. Front Bioeng Biotechnol 2020; 8:595497. [PMID: 33240867 PMCID: PMC7680962 DOI: 10.3389/fbioe.2020.595497] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Extensive studies have shown that cells can sense and modulate the biomechanical properties of the ECM within their resident microenvironment. Thus, targeting the mechanotransduction signaling pathways provides a promising way for disease intervention. However, how cells perceive these mechanical cues of the microenvironment and transduce them into biochemical signals remains to be answered. Förster or fluorescence resonance energy transfer (FRET) based biosensors are a powerful tool that can be used in live-cell mechanotransduction imaging and mechanopharmacological drug screening. In this review, we will first introduce FRET principle and FRET biosensors, and then, recent advances on the integration of FRET biosensors and mechanobiology in normal and pathophysiological conditions will be discussed. Furthermore, we will summarize the current applications and limitations of FRET biosensors in high-throughput drug screening and the future improvement of FRET biosensors. In summary, FRET biosensors have provided a powerful tool for mechanobiology studies to advance our understanding of how cells and matrices interact, and the mechanopharmacological screening for disease intervention.
Collapse
Affiliation(s)
| | | | | | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
5
|
Agarwal G, Gabrani R. Antiviral Peptides: Identification and Validation. Int J Pept Res Ther 2020; 27:149-168. [PMID: 32427225 PMCID: PMC7233194 DOI: 10.1007/s10989-020-10072-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/30/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022]
Abstract
Despite rapid advances in the human healthcare, the infection caused by certain viruses results in high morbidity and mortality accentuate the importance for development of new antivirals. The existing antiviral drugs are limited, due to their inadequate response, increased rate of resistance and several adverse side effects. Therefore, one of the newly emerging field “peptide-based therapeutics” against viruses is being explored and seems promising. Over the last few years, a lot of scientific effort has been made for the identification of novel and potential peptide-based therapeutics using various advanced technologies. Consequently, there are more than 60 approved peptide drugs available for sale in the market of United States, Europe, Japan, and some Asian countries. Moreover, the number of peptide drugs undergoing the clinical trials is rising gradually year by year. The peptide-based antiviral therapeutics have been approved for the Human immunodeficiency virus (HIV), Influenza virus and Hepatitis virus (B and C). This review enlightens the various peptide sources and the different approaches that have contributed to the search of potential antiviral peptides. These include computational approaches, natural and biological sources (library based high throughput screening) for the identification of lead peptide molecules against their target. Further the applications of few advanced techniques based on combinatorial chemistry and molecular biology have been illustrated to measure the binding parameters such as affinity and kinetics of the screened interacting partners. The employment of these advanced techniques can contribute to investigate antiviral peptide therapeutics for emerging infections.
Collapse
Affiliation(s)
- Garima Agarwal
- Department of Biotechnology, Center for Emerging Diseases, Jaypee Institute of Information Technology, Noida, UP 201309 India
| | - Reema Gabrani
- Department of Biotechnology, Center for Emerging Diseases, Jaypee Institute of Information Technology, Noida, UP 201309 India
| |
Collapse
|
6
|
Senarisoy M, Barette C, Lacroix F, De Bonis S, Stelter M, Hans F, Kleman JP, Fauvarque MO, Timmins J. Förster Resonance Energy Transfer Based Biosensor for Targeting the hNTH1-YB1 Interface as a Potential Anticancer Drug Target. ACS Chem Biol 2020; 15:990-1003. [PMID: 32125823 DOI: 10.1021/acschembio.9b01023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Y-box binding protein 1 (YB1) is an established metastatic marker: high expression and nuclear localization of YB1 correlate with tumor aggressiveness, drug resistance, and poor patient survival in various tumors. In the nucleus, YB1 interacts with and regulates the activities of several nuclear proteins, including the DNA glycosylase, human endonuclease III (hNTH1). In the present study, we used Förster resonance energy transfer (FRET) and AlphaLISA technologies to further characterize this interaction and define the minimal regions of hNTH1 and YB1 required for complex formation. This work led us to design an original and cost-effective FRET-based biosensor for the rapid in vitro high-throughput screening for potential inhibitors of the hNTH1-YB1 complex. Two pilot screens were carried out, allowing the selection of several promising compounds exhibiting IC50 values in the low micromolar range. Interestingly, two of these compounds bind to YB1 and sensitize drug-resistant breast tumor cells to the chemotherapeutic agent, cisplatin. Taken together, these findings demonstrate that the hNTH1-YB1 interface is a druggable target for the development of new therapeutic strategies for the treatment of drug-resistant tumors. Moreover, beyond this study, the simple design of our biosensor defines an innovative and efficient strategy for the screening of inhibitors of therapeutically relevant protein-protein interfaces.
Collapse
Affiliation(s)
- Muge Senarisoy
- Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Caroline Barette
- Univ. Grenoble Alpes, CEA, INSERM, BGE, F-38000 Grenoble, France
| | | | | | - Meike Stelter
- Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Fabienne Hans
- Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | | | | | - Joanna Timmins
- Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| |
Collapse
|
7
|
Finnell JG, Tsang TM, Cryan L, Garrard S, Lee SL, Ackroyd PC, Rogers MS, Christensen KA. A Canstatin-Derived Peptide Provides Insight into the Role of Capillary Morphogenesis Gene 2 in Angiogenic Regulation and Matrix Uptake. ACS Chem Biol 2020; 15:587-596. [PMID: 32003961 DOI: 10.1021/acschembio.0c00064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Capillary Morphogenesis Gene 2 protein (CMG2) is a transmembrane, integrin-like receptor and the primary receptor for the anthrax toxin. CMG2 also plays a role in angiogenic processes. However, the molecular mechanism that mediates the observed CMG2-related angiogenic effects is not fully elucidated. Previous studies have reported that CMG2 binds type IV collagen (Col-IV), a vital component of the vascular basement membrane, as well as other ECM proteins. Here, we further characterize the interaction between CMG2 and individual peptides from Col-IV and explore the effects of this interaction on angiogenesis. Using a peptide array, we observed that CMG2 preferentially binds peptide fragments of the NC1 (noncollagenous domain 1) domains of Col-IV. These domains are also known as the fragments arresten (from the α1 chain) and canstatin (from the α2 chain) and have documented antiangiogenic properties. A second peptide array was probed to map a putative peptide-binding epitope onto the Col-IV structure. A top hit from the initial array, a canstatin-derived peptide, binds to the CMG2 ligand-binding von Willebrand factor A (vWA) domain with a submicromolar affinity (peptide S16, Kd = 400 ± 200 nM). This peptide competes with anthrax protective antigen (PA) for CMG2 binding and does not bind CMG2 in the presence of EDTA. Together these data suggest that, like PA, S16 interacts with CMG2 at the metal-ion dependent adhesion site (MIDAS) of its vWA domain. CMG2 specifically mediates endocytic uptake of S16; both CMG2-/- endothelial cells and WT cells treated with PA show markedly reduced S16 uptake. Furthermore, S16 dramatically reduces directional endothelial cell migration with no impact on cell proliferation. These data demonstrate that this canstatin-derived peptide acts via CMG2 to elicit a marked effect on a critical process required for angiogenesis.
Collapse
Affiliation(s)
- Jordan G. Finnell
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Tsz-Ming Tsang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Lorna Cryan
- Vascular Biology Program, Boston Children’s Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Samuel Garrard
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Sai-Lun Lee
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - P. Christine Ackroyd
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Michael S. Rogers
- Vascular Biology Program, Boston Children’s Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Kenneth A. Christensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| |
Collapse
|
8
|
Klisara N, Yu YM, Palaniappan A, Liedberg B. Towards on-site visual detection of proteases in food matrices. Anal Chim Acta 2019; 1078:182-188. [DOI: 10.1016/j.aca.2019.06.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/22/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022]
|
9
|
G.-Doyagüez E, Carrero P, Madrona A, Rodriguez-Salamanca P, Martínez-Gualda B, Camarasa MJ, Jimeno ML, Bennallack PR, Finnell JG, Tsang TM, Christensen KA, San-Félix A, Rogers MS. Galloyl Carbohydrates with Antiangiogenic Activity Mediated by Capillary Morphogenesis Gene 2 (CMG2) Protein Binding. J Med Chem 2019; 62:3958-3970. [DOI: 10.1021/acs.jmedchem.8b01988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Elisa G.-Doyagüez
- Instituto de Química Médica (IQM, CSIC), 28006 Madrid, Spain
- Centro de Química Orgánica “Lora-Tamayo” (CENQUIOR, CSIC), 28006 Madrid, Spain
| | - Paula Carrero
- Instituto de Química Médica (IQM, CSIC), 28006 Madrid, Spain
| | - Andrés Madrona
- Instituto de Química Médica (IQM, CSIC), 28006 Madrid, Spain
| | | | | | | | - María Luisa Jimeno
- Centro de Química Orgánica “Lora-Tamayo” (CENQUIOR, CSIC), 28006 Madrid, Spain
| | - Philip R. Bennallack
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jordan G. Finnell
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Tsz-Ming Tsang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Kenneth A. Christensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah 84602, United States
| | - Ana San-Félix
- Instituto de Química Médica (IQM, CSIC), 28006 Madrid, Spain
| | - Michael S. Rogers
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
10
|
Liu W, Cui Y, Ren W, Irudayaraj J. Epigenetic biomarker screening by FLIM-FRET for combination therapy in ER+ breast cancer. Clin Epigenetics 2019; 11:16. [PMID: 30700309 PMCID: PMC6354376 DOI: 10.1186/s13148-019-0620-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/21/2019] [Indexed: 01/13/2023] Open
Abstract
Hormone-dependent gene expression involves dynamic and orchestrated regulation of epigenome leading to a cancerous state. Estrogen receptor (ER)-positive breast cancer rely on chromatin remodeling and association with epigenetic factors in inducing ER-dependent oncogenesis and thus cell over-proliferation. The mechanistic differences between epigenetic regulation and hormone signaling provide an avenue for combination therapy of ER-positive breast cancer. We hypothesized that epigenetic biomarkers within single nucleosome proximity of ER-dependent genes could serve as potential therapeutic targets. We described here a Fluorescence lifetime imaging-based Förster resonance energy transfer (FLIM-FRET) methodology for biomarker screening that could facilitate combination therapy based on our study. We screened 11 epigenetic-related markers which include oxidative forms of DNA methylation, histone modifications, and methyl-binding domain proteins. Among them, we identified H4K12acetylation (H4K12ac) and H3K27 acetylation (H3K27ac) as potential epigenetic therapeutic targets. When histone acetyltransferase inhibitor targeting H4K12ac and H3K27ac was combined with tamoxifen, an enhanced therapeutic outcome was observed against ER-positive breast cancer both in vitro and in vivo. Together, we demonstrate a single molecule approach as an effective screening tool for devising targeted epigenetic therapy.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
- Present Address: Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA
| | - Yi Cui
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
- Earth and Biological Science Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Wen Ren
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
| | - Joseph Irudayaraj
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA.
- Present Address: Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA.
| |
Collapse
|
11
|
Capillary morphogenesis gene 2 maintains gastric cancer stem-like cell phenotype by activating a Wnt/β-catenin pathway. Oncogene 2018; 37:3953-3966. [PMID: 29662192 PMCID: PMC6053357 DOI: 10.1038/s41388-018-0226-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/29/2017] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
Abstract
A growing body of evidence shows that the development and progression of gastric cancer (GC) is mainly associated to the presence of gastric cancer stem-like cells (GCSLCs). However, it is unclear how GCSLC population is maintained. This study aimed to explore the role of capillary morphogenesis gene 2 (CMG2) in GCSLC maintenance and the relevance to GC progression. We found that CMG2 was highly expressed in GC tissues and the expression levels were associated with the invasion depth and lymph node metastasis of GC, and inversely correlated with the survival of GC patients. Sorted CMG2High GC cells preferentially clustered in CD44High stem-like cell population, which expressed high levels of stemness-related genes with increased capabilities of self-renewal and tumorigenicity. Depletion of CMG2 gene resulted in reduction of GCSLC population with attenuated stemness and decrease of invasive and metastatic capabilities with subdued epithelial–mesenchymal transition phenotype in GC cells. Mechanistically, CMG2 interacted with LRP6 in GCSLCs to activate a Wnt/β-catenin pathway. Thus, our results demonstrate that CMG2 promotes GC progression by maintaining GCSLCs and can serve as a new prognostic indicator and a target for human GC therapy.
Collapse
|
12
|
Huang BX, Newcomer K, Kevala K, Barnaeva E, Zheng W, Hu X, Patnaik S, Southall N, Marugan J, Ferrer M, Kim HY. Identification of 4-phenylquinolin-2(1H)-one as a specific allosteric inhibitor of Akt. Sci Rep 2017; 7:11673. [PMID: 28916818 PMCID: PMC5601486 DOI: 10.1038/s41598-017-11870-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/31/2017] [Indexed: 12/28/2022] Open
Abstract
Akt plays a major role in tumorigenesis and the development of specific Akt inhibitors as effective cancer therapeutics has been challenging. Here, we report the identification of a highly specific allosteric inhibitor of Akt through a FRET-based high-throughput screening, and characterization of its inhibitory mechanism. Out of 373,868 compounds screened, 4-phenylquinolin-2(1H)-one specifically decreased Akt phosphorylation at both T308 and S473, and inhibited Akt kinase activity (IC50 = 6 µM) and downstream signaling. 4-Phenylquinolin-2(1H)-one did not alter the activity of upstream kinases including PI3K, PDK1, and mTORC2 as well as closely related kinases that affect cell proliferation and survival such as SGK1, PKA, PKC, or ERK1/2. This compound inhibited the proliferation of cancer cells but displayed less toxicity compared to inhibitors of PI3K or mTOR. Kinase profiling efforts revealed that 4-phenylquinolin-2(1H)-one does not bind to the kinase active site of over 380 human kinases including Akt. However, 4-phenylquinolin-2(1H)-one interacted with the PH domain of Akt, apparently inducing a conformation that hinders S473 and T308 phosphorylation by mTORC2 and PDK1. In conclusion, we demonstrate that 4-phenylquinolin-2(1H)-one is an exquisitely selective Akt inhibitor with a distinctive molecular mechanism, and a promising lead compound for further optimization toward the development of novel cancer therapeutics.
Collapse
Affiliation(s)
- Bill X Huang
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, NIH, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Kenny Newcomer
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, NIH, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Karl Kevala
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, NIH, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Elena Barnaeva
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Wei Zheng
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Xin Hu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Samarjit Patnaik
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Noel Southall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Juan Marugan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, NIH, 5625 Fishers Lane, Rockville, MD, 20852, USA.
| |
Collapse
|
13
|
Targeting Bacillus anthracis toxicity with a genetically selected inhibitor of the PA/CMG2 protein-protein interaction. Sci Rep 2017; 7:3104. [PMID: 28596569 PMCID: PMC5465072 DOI: 10.1038/s41598-017-03253-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/25/2017] [Indexed: 11/23/2022] Open
Abstract
The protein-protein interaction between the human CMG2 receptor and the Bacillus anthracis protective antigen (PA) is essential for the transport of anthrax lethal and edema toxins into human cells. We used a genetically encoded high throughput screening platform to screen a SICLOPPS library of 3.2 million cyclic hexapeptides for inhibitors of this protein-protein interaction. Unusually, the top 3 hits all contained stop codons in the randomized region of the library, resulting in linear rather than cyclic peptides. These peptides disrupted the targeted interaction in vitro; two act by binding to CMG2 while one binds PA. The efficacy of the most potent CMG2-binding inhibitor was improved through the incorporation of non-natural phenylalanine analogues. Cell based assays demonstrated that the optimized inhibitor protects macrophages from the toxicity of lethal factor.
Collapse
|
14
|
Fluorescence Resonance Energy Transfer Microscopy for Measuring Chromatin Complex Structure and Dynamics. Methods Mol Biol 2016. [PMID: 27659982 DOI: 10.1007/978-1-4939-6380-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The Polycomb group (PcG) proteins form regulatory complexes that modify the chromatin structure and silence their target genes. Recent works have found that the composition of Polycomb complexes is highly dynamic. Defining the different protein components of each complex is fundamental for better understanding their biological functions. Fluorescent resonance energy transfer (FRET) is a powerful tool to measure protein-protein interactions, in nanometer order and in their native cellular environment. Here we describe the preparation and execution of a typical FRET experiment using CFP-tagged protein as donor and YFP-tagged protein as acceptor. We further show that FRET can be used in a competition assay to measure binding affinities of different components of the same chromatin complex.
Collapse
|
15
|
Ohanjanian L, Remy KE, Li Y, Cui X, Eichacker PQ. An overview of investigational toxin-directed therapies for the adjunctive management of Bacillus anthracis infection and sepsis. Expert Opin Investig Drugs 2015; 24:851-65. [PMID: 25920540 DOI: 10.1517/13543784.2015.1041587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sepsis with Bacillus anthracis infection has a very high mortality rate despite appropriate antibiotic and supportive therapies. Over the past 15 years, recent outbreaks in the US and in Europe, coupled with anthrax's bioterrorism weapon potential, have stimulated efforts to develop adjunctive therapies to improve clinical outcomes. Since lethal toxin and edema toxin (LT and ET) make central contributions to the pathogenesis of B. anthracis, these have been major targets in this effort. AREAS COVERED Here, the authors review different investigative biopharmaceuticals that have been recently identified for their therapeutic potential as inhibitors of LT or ET. Among these inhibitors are two antibody preparations that have been included in the Strategic National Stockpile (SNS) and several more that have reached Phase I testing. Presently, however, many of these candidate agents have only been studied in vitro and very few tested in bacteria-challenged models. EXPERT OPINION Although a large number of drugs have been identified as potential therapeutic inhibitors of LT and ET, in most cases their testing has been limited. The use of the two SNS antibody therapies during a large-scale exposure to B. anthracis will be difficult. Further testing and development of agents with oral bioavailability and relatively long shelf lives should be a focus for future research.
Collapse
Affiliation(s)
- Lernik Ohanjanian
- National Institutes of Health, Clinical Center, Critical Care Medicine Department , Building 10, Room 2C145, Bethesda, MD 20892 , USA +1 301 402 2914 ; +1 301 402 1213 ;
| | | | | | | | | |
Collapse
|
16
|
Abstract
Resistance of important bacterial pathogens to common antimicrobial therapies and the emergence of multidrug-resistant bacteria are increasing at an alarming rate and constitute one of our greatest challenges in the combat of bacterial infection and accompanied diseases. The current shortage of effective drugs, lack of successful prevention measures and only a few new antibiotics in the clinical pipeline demand the development of novel treatment options and alternative antimicrobial therapies. Our increasing understanding of bacterial virulence strategies and the induced molecular pathways of the infectious disease provides novel opportunities to target and interfere with crucial pathogenicity factors or virulence-associated traits of the bacteria while bypassing the evolutionary pressure on the bacterium to develop resistance. In the past decade, numerous new bacterial targets for anti-virulence therapies have been identified, and structure-based tailoring of intervention strategies and screening assays for small-molecule inhibitors of such pathways were successfully established. In this chapter, we will take a closer look at the bacterial virulence-related factors and processes that present promising targets for anti-virulence therapies, recently discovered inhibitory substances and their promises and discuss the challenges, and problems that have to be faced.
Collapse
|
17
|
Angiogenic growth factors interactome and drug discovery: The contribution of surface plasmon resonance. Cytokine Growth Factor Rev 2014; 26:293-310. [PMID: 25465594 DOI: 10.1016/j.cytogfr.2014.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 11/21/2022]
Abstract
Angiogenesis is implicated in several pathological conditions, including cancer, and in regenerative processes, including the formation of collateral blood vessels after stroke. Physiological angiogenesis is the outcome of a fine balance between the action of angiogenic growth factors (AGFs) and anti-angiogenic molecules, while pathological angiogenesis occurs when this balance is pushed toward AGFs. AGFs interact with multiple endothelial cell (EC) surface receptors inducing cell proliferation, migration and proteases upregulation. On the contrary, free or extracellular matrix-associated molecules inhibit angiogenesis by sequestering AGFs (thus hampering EC stimulation) or by interacting with specific EC receptors inducing apoptosis or decreasing responsiveness to AGFs. Thus, angiogenesis results from an intricate network of interactions among pro- and anti-angiogenic molecules, EC receptors and various modulators. All these interactions represent targets for the development of pro- or anti-angiogenic therapies. These aims call for suitable technologies to study the countless interactions occurring during neovascularization. Surface plasmon resonance (SPR) is a label-free optical technique to study biomolecular interactions in real time. It has become the golden standard technology for interaction analysis in biomedical research, including angiogenesis. From a survey of the literature it emerges that SPR has already contributed substantially to the better understanding of the neovascularization process, laying the basis for the decoding of the angiogenesis "interactome" and the identification of "hub molecules" that may represent preferential targets for an efficacious modulation of angiogenesis. Here, the still unexploited full potential of SPR is enlightened, pointing to improvements in its use for a deeper understanding of the mechanisms of neovascularization and the identification of novel anti-angiogenic drugs.
Collapse
|
18
|
Ye L, Sun PH, Sanders AJ, Martin TA, Lane J, Mason MD, Jiang WG. Therapeutic potential of capillary morphogenesis gene 2 extracellular vWA domain in tumour‑related angiogenesis. Int J Oncol 2014; 45:1565-73. [PMID: 24993339 DOI: 10.3892/ijo.2014.2533] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/14/2014] [Indexed: 11/05/2022] Open
Abstract
Capillary morphogenesis gene 2 (CMG2) is a receptor of anthrax toxin and plays an important role in angiogenesis. It has been shown to be involved in the cell adhesion and motility of various cell types, including epithelia and endothelia. The present study aimed to examine the therapeutic potential of targeting CMG2 to prevent tumour‑related new vasculature. The full-length coding sequence of the human CMG2 gene and different fragments of the CMG2 vWA domain were amplified and constructed into a mammalian expression plasmid vector. The effect of CMG2 and its vWA domain on endothelial cells and angiogenesis was assessed using relevant in vitro, ex vivo and in vivo models. The overexpression of CMG2 enhanced the adhesion of endothelial cells to extracellular matrix, but was negatively associated with cell migration. Overexpression of CMG2 and the vWA domain fragments inhibited the tubule formation and migration of endothelial cells. Small peptides based on the amino acid sequence of the CMG2 vWA domain fragments potently inhibited in vitro tubule formation and ex vivo angiogenesis. One of the polypeptides, LG20, showed an inhibitory effect on in vivo tumour growth of cancer cells which were co-inoculated with the vascular endothelial cells. CMG2 is a potential target for treating tumour‑related angiogenesis. The polypeptides based on the CMG2 vWA domain can potently inhibit in vitro and ex vivo angiogenesis, which may contribute to the inhibitory effect on in vivo tumour growth. Further investigations are required to shed light on the machinery and may provide a novel therapeutic approach for inhibition of angiogenesis in cancer management.
Collapse
Affiliation(s)
- Lin Ye
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Ping-Hui Sun
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew J Sanders
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Tracey A Martin
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Jane Lane
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Malcolm D Mason
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Wen G Jiang
- Metastasis and Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
19
|
Abstract
INTRODUCTION Present-day rational drug design approaches are based on exploiting unique features of the target biomolecules, small- or macromolecule drug candidates and physical forces that govern their interactions. The 2013 Nobel Prize in chemistry awarded 'for the development of multiscale models for complex chemical systems' once again demonstrated the importance of the tailored drug discovery that reduces the role of the trial-and-error approach to a minimum. The intentional dissemination of Bacillus anthracis spores in 2001 via the so-called anthrax letters has led to increased efforts, politically and scientifically, to develop medical countermeasures that will protect people from the threat of anthrax bioterrorism. AREAS COVERED This article provides an overview of the recent rational drug design approaches for discovering inhibitors of anthrax toxin. The review also directs the readers to the vast literature on the recognized advances and future possibilities in the field. EXPERT OPINION Existing options to combat anthrax toxin lethality are limited. With the only anthrax toxin inhibiting therapy (protective antigen-targeting with a monoclonal antibody, raxibacumab) approved to treat inhalational anthrax, the situation, in our view, is still insecure. Further, the FDA's animal rule for drug approval, which clears compounds without validated efficacy studies on humans, creates a high level of uncertainty, especially when a well-characterized animal model does not exist. Better identification and validation of anthrax toxin therapeutic targets at the molecular level as well as elucidation of the parameters determining the corresponding therapeutic windows are still necessary for more effective therapeutic options.
Collapse
Affiliation(s)
- Ekaterina M Nestorovich
- The Catholic University of America, Department of Biology , Washington, DC , USA +1 202 319 6723 ;
| | | |
Collapse
|
20
|
Cryan LM, Bazinet L, Habeshian KA, Cao S, Clardy J, Christensen KA, Rogers MS. 1,2,3,4,6-Penta-O-galloyl-β-D-glucopyranose inhibits angiogenesis via inhibition of capillary morphogenesis gene 2. J Med Chem 2013; 56:1940-5. [PMID: 23394144 PMCID: PMC3600088 DOI: 10.1021/jm301558t] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Capillary morphogenesis gene 2 (CMG2) is a transmembrane extracellular matrix binding protein that is also an anthrax toxin receptor. We have shown that high-affinity CMG2 binders can inhibit angiogenesis and tumor growth. We recently described a high-throughput FRET assay to identify CMG2 inhibitors. We now report the serendipitous discovery that PGG (1,2,3,4,6-penta-O-galloyl-β-D-glucopyranose) is a CMG2 inhibitor with antiangiogenic activity. PGG is a gallotannin produced by a variety of medicinal plants that exhibits a wide variety of antitumor and other activities. We find that PGG inhibits CMG2 with a submicromolar IC50 and it also inhibits the migration of human dermal microvascular endothelial cells at similar concentrations in vitro. Finally, oral or intraperitoneal administration of PGG inhibits angiogenesis in the mouse corneal micropocket assay in vivo. Together, these results suggest that a portion of the in vivo antitumor activity of PGG may be the result of antiangiogenic activity mediated by inhibition of CMG2.
Collapse
Affiliation(s)
- Lorna M. Cryan
- Vascular Biology Program, Department of Surgery, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Lauren Bazinet
- Vascular Biology Program, Department of Surgery, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Kaiane A. Habeshian
- Vascular Biology Program, Department of Surgery, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| | - Shugeng Cao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115
| | | | - Michael S. Rogers
- Vascular Biology Program, Department of Surgery, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
21
|
Cryan LM, Habeshian KA, Caldwell TP, Morris MT, Ackroyd PC, Christensen KA, Rogers MS. Identification of small molecules that inhibit the interaction of TEM8 with anthrax protective antigen using a FRET assay. ACTA ACUST UNITED AC 2013; 18:714-25. [PMID: 23479355 DOI: 10.1177/1087057113478655] [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] [Indexed: 11/15/2022]
Abstract
Tumor marker endothelial 8 (TEM8) is a receptor for the protective antigen (PA) component of anthrax toxin. TEM8 is upregulated on endothelial cells lining the blood vessels within tumors, compared with normal blood vessels. A number of studies have demonstrated a pivotal role for TEM8 in developmental and tumor angiogenesis. We have also shown that targeting the anthrax receptors with a mutated form of PA inhibits angiogenesis and tumor formation in vivo. Here we describe the development and testing of a high-throughput fluorescence resonance energy transfer assay to identify molecules that strongly inhibit the interaction of PA and TEM8. The assay we describe is sensitive and robust, with a Z' value of 0.8. A preliminary screen of 2310 known bioactive library compounds identified ebselen and thimerosal as inhibitors of the TEM8-PA interaction. These molecules each contain a cysteine-reactive transition metal, and complementary studies indicate that their inhibition of interaction is due to modification of a cysteine residue in the TEM8 extracellular domain. This is the first demonstration of a high-throughput screening assay that identifies inhibitors of TEM8, with potential application for antianthrax and antiangiogenic diseases.
Collapse
Affiliation(s)
- Lorna M Cryan
- Boston Children’s Hospital, Harvard Medical School, Vascular Biology Program, Department of Surgery, Karp 11, 300 Longwood Ave, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|