1
|
Powell J, Mann CA, Toth PD, Nolan S, Steinert A, Ove C, Seffernick JT, Wozniak DJ, Kebriaei R, Lindert S, Osheroff N, Yalowich JC, Mitton-Fry MJ. Development of Novel Bacterial Topoisomerase Inhibitors Assisted by Computational Screening. ACS Med Chem Lett 2024; 15:1287-1297. [PMID: 39140037 PMCID: PMC11318591 DOI: 10.1021/acsmedchemlett.4c00162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 08/15/2024] Open
Abstract
Multidrug-resistant bacterial infections pose an ever-evolving threat to public health. Since the outset of the antibacterial age, bacteria have developed a multitude of diverse resistance mechanisms that suppress the effectiveness of current therapies. New drug entities, such as Novel Bacterial Topoisomerase Inhibitors (NBTIs), can circumvent this major issue. A computational docking model was employed to predict the binding to DNA gyrase of atypical NBTIs with novel pharmacophores. Synthesis of NBTIs based on computational docking and subsequent antibacterial evaluation against both Gram-positive and Gram-negative bacteria yielded congeners with outstanding anti-staphylococcal activity and varying activity against select Gram-negative pathogens.
Collapse
Affiliation(s)
- Joshua
W. Powell
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chelsea A. Mann
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Paul D. Toth
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Sheri Nolan
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Anja Steinert
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Clarissa Ove
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Justin T. Seffernick
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Daniel J. Wozniak
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Razieh Kebriaei
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Steffen Lindert
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Neil Osheroff
- Department of Biochemistry and Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jack C. Yalowich
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Mark J. Mitton-Fry
- Division
of Medicinal Chemistry and Pharmacognosy, Department of Chemistry and Biochemistry, Microbial Infection
and Immunity, Division of Outcomes and Translational Sciences, Department of Microbiology, and Division of Pharmaceutics
and Pharmacology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
2
|
Jung JW, Park SY, Seo EJ, Jang IH, Park Y, Lee D, Kim D, Kim JM. Functional expression of oxytocin receptors in pulp-dentin complex. Biomaterials 2023; 293:121977. [PMID: 36580714 DOI: 10.1016/j.biomaterials.2022.121977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/24/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Dental pulp-derived stromal cells (DPSCs) are a crucial cell population for maintaining the tissue integrity of the pulp-dentin complex. The oxytocin receptor (OXTR), a member of the G protein-coupled receptor (GPCR) superfamily, plays versatile roles in diverse biological contexts. However, the role of OXTR in dental pulp has not yet been fully understood. Here, we demonstrate the biological functions and significance of OXTR in DPSCs through a multidisciplinary approach. Microarray data of 494 GPCR genes revealed high OXTR expression in human DPSCs (hDPSCs). Blocking OXTR activity increased the expression of osteogenic and odontogenic marker genes, promoting hDPSC differentiation. Additionally, we found that OXTR is involved in extracellular matrix (ECM) remodeling through the regulation of the gene expression related to ECM homeostasis. We further demonstrated that these genetic changes are mediated by trascriptional activity of Yes-associated protein (YAP). Based on the results, a preclinical experiment was performed using an animal model, demonstrating that the application of an OXTR inhibitor to damaged pulp induced significant hard tissue formation. These results provide new insight into the oxytocin-OXTR system in the regenerative process of pulp-dentin complex.
Collapse
Affiliation(s)
- Ju Won Jung
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - So Young Park
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun Jin Seo
- Department of Oral Biochemistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Il Ho Jang
- Department of Oral Biochemistry, Pusan National University, Yangsan, 50612, Republic of Korea; Dental and Life Science Institute, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Yeji Park
- Department of Conservative Dentistry and Oral Science Research Center, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Dasun Lee
- Department of Conservative Dentistry and Oral Science Research Center, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Dohyun Kim
- Department of Conservative Dentistry and Oral Science Research Center, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea.
| | - Jin Man Kim
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
3
|
Luo M, Xu Y, Chen H, Wu Y, Pang A, Hu J, Dong X, Che J, Yang H. Advances of targeting the YAP/TAZ-TEAD complex in the hippo pathway for the treatment of cancers. Eur J Med Chem 2022; 244:114847. [DOI: 10.1016/j.ejmech.2022.114847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/03/2022]
|
4
|
Gao Z, Fan T, Chen L, Yang M, Wai Wong VK, Chen D, Liu Z, Zhou Y, Wu W, Qiu Z, Zhang C, Li Y, Jiang Y. Design, synthesis and antitumor evaluation of novel 1H-indole-2-carboxylic acid derivatives targeting 14-3-3η protein. Eur J Med Chem 2022; 238:114402. [DOI: 10.1016/j.ejmech.2022.114402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/06/2022] [Accepted: 04/17/2022] [Indexed: 11/04/2022]
|
5
|
Andl T, Andl CD, Zhang Y. Two-edged sword: how activation of the "proto-oncogene" yes-associated protein 1 in lung squamous cell carcinoma can surprisingly inhibit tumor growth. J Thorac Dis 2019; 10:S3870-S3874. [PMID: 30631502 DOI: 10.21037/jtd.2018.10.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Thomas Andl
- Burnett School of Biological Sciences, University of Central Florida, Orlando, FL, USA
| | - Claudia D Andl
- Burnett School of Biological Sciences, University of Central Florida, Orlando, FL, USA
| | - Yuhang Zhang
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
6
|
Leanza L, Checchetto V, Biasutto L, Rossa A, Costa R, Bachmann M, Zoratti M, Szabo I. Pharmacological modulation of mitochondrial ion channels. Br J Pharmacol 2019; 176:4258-4283. [PMID: 30440086 DOI: 10.1111/bph.14544] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022] Open
Abstract
The field of mitochondrial ion channels has undergone a rapid development during the last three decades, due to the molecular identification of some of the channels residing in the outer and inner membranes. Relevant information about the function of these channels in physiological and pathological settings was gained thanks to genetic models for a few, mitochondria-specific channels. However, many ion channels have multiple localizations within the cell, hampering a clear-cut determination of their function by pharmacological means. The present review summarizes our current knowledge about the ins and outs of mitochondrial ion channels, with special focus on the channels that have received much attention in recent years, namely, the voltage-dependent anion channels, the permeability transition pore (also called mitochondrial megachannel), the mitochondrial calcium uniporter and some of the inner membrane-located potassium channels. In addition, possible strategies to overcome the difficulties of specifically targeting mitochondrial channels versus their counterparts active in other membranes are discussed, as well as the possibilities of modulating channel function by small peptides that compete for binding with protein interacting partners. Altogether, these promising tools along with large-scale chemical screenings set up to identify new, specific channel modulators will hopefully allow us to pinpoint the actual function of most mitochondrial ion channels in the near future and to pharmacologically affect important pathologies in which they are involved, such as neurodegeneration, ischaemic damage and cancer. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Andrea Rossa
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Roberto Costa
- Department of Biology, University of Padova, Padova, Italy
| | | | - Mario Zoratti
- CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
7
|
Zaccagnino A, Managò A, Leanza L, Gontarewitz A, Linder B, Azzolini M, Biasutto L, Zoratti M, Peruzzo R, Legler K, Trauzold A, Kalthoff H, Szabo I. Tumor-reducing effect of the clinically used drug clofazimine in a SCID mouse model of pancreatic ductal adenocarcinoma. Oncotarget 2018; 8:38276-38293. [PMID: 27542263 PMCID: PMC5503532 DOI: 10.18632/oncotarget.11299] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/09/2016] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the most common form of pancreatic cancer with rising incidence in developing countries. Unfortunately, the overall 5-year survival rate is still less than 5%. The most frequent oncogenic mutations in PDAC are loss-of function mutations in p53 and gain-of-function mutations in KRAS. Here we show that clofazimine (Lamprene), a drug already used in the clinic for autoimmune diseases and leprosy, is able to efficiently kill in vitro five different PDAC cell lines harboring p53 mutations. We provide evidence that clofazimine induces apoptosis in PDAC cells with an EC50 in the μM range via its specific inhibitory action on the potassium channel Kv1.3. Intraperitoneal injection of clofazimine resulted in its accumulation in the pancreas of mice 8 hours after administration. Using an orthotopic PDAC xenotransplantation model in SCID beige mouse, we show that clofazimine significantly and strongly reduced the primary tumor weight. Thus, our work identifies clofazimine as a promising therapeutic agent against PDAC and further highlights ion channels as possible oncological targets.
Collapse
Affiliation(s)
- Angela Zaccagnino
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, Arnold-Heller-Strasse 3 (Haus 17) D-24105 Kiel, Germany
| | - Antonella Managò
- Department of Biology, University of Padova, viale G. Colombo 3. Padova, I-35121 Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, viale G. Colombo 3. Padova, I-35121 Italy
| | - Artur Gontarewitz
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, Arnold-Heller-Strasse 3 (Haus 17) D-24105 Kiel, Germany
| | - Bernhard Linder
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, Arnold-Heller-Strasse 3 (Haus 17) D-24105 Kiel, Germany
| | - Michele Azzolini
- Department of Biomedical Sciences, University of Padova, I-35121 Italy.,CNR Institute of Neuroscience, Padova, Italy
| | - Lucia Biasutto
- Department of Biomedical Sciences, University of Padova, I-35121 Italy.,CNR Institute of Neuroscience, Padova, Italy
| | - Mario Zoratti
- Department of Biomedical Sciences, University of Padova, I-35121 Italy.,CNR Institute of Neuroscience, Padova, Italy
| | - Roberta Peruzzo
- Department of Biology, University of Padova, viale G. Colombo 3. Padova, I-35121 Italy
| | - Karen Legler
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, Arnold-Heller-Strasse 3 (Haus 17) D-24105 Kiel, Germany
| | - Anna Trauzold
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, Arnold-Heller-Strasse 3 (Haus 17) D-24105 Kiel, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, Arnold-Heller-Strasse 3 (Haus 17) D-24105 Kiel, Germany
| | - Ildiko Szabo
- Department of Biology, University of Padova, viale G. Colombo 3. Padova, I-35121 Italy.,CNR Institute of Neuroscience, Padova, Italy
| |
Collapse
|
8
|
Calhoun S, Korczynska M, Wichelecki DJ, San Francisco B, Zhao S, Rodionov DA, Vetting MW, Al-Obaidi NF, Lin H, O'Meara MJ, Scott DA, Morris JH, Russel D, Almo SC, Osterman AL, Gerlt JA, Jacobson MP, Shoichet BK, Sali A. Prediction of enzymatic pathways by integrative pathway mapping. eLife 2018; 7:31097. [PMID: 29377793 PMCID: PMC5788505 DOI: 10.7554/elife.31097] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/18/2017] [Indexed: 01/17/2023] Open
Abstract
The functions of most proteins are yet to be determined. The function of an enzyme is often defined by its interacting partners, including its substrate and product, and its role in larger metabolic networks. Here, we describe a computational method that predicts the functions of orphan enzymes by organizing them into a linear metabolic pathway. Given candidate enzyme and metabolite pathway members, this aim is achieved by finding those pathways that satisfy structural and network restraints implied by varied input information, including that from virtual screening, chemoinformatics, genomic context analysis, and ligand -binding experiments. We demonstrate this integrative pathway mapping method by predicting the L-gulonate catabolic pathway in Haemophilus influenzae Rd KW20. The prediction was subsequently validated experimentally by enzymology, crystallography, and metabolomics. Integrative pathway mapping by satisfaction of structural and network restraints is extensible to molecular networks in general and thus formally bridges the gap between structural biology and systems biology.
Collapse
Affiliation(s)
- Sara Calhoun
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Magdalena Korczynska
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Daniel J Wichelecki
- Institute for Genomic Biology, University of Illinois, Urbana, United States.,Department of Biochemistry, University of Illinois, Urbana, United States.,Department of Chemistry, University of Illinois, Urbana, United States
| | - Brian San Francisco
- Institute for Genomic Biology, University of Illinois, Urbana, United States
| | - Suwen Zhao
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Dmitry A Rodionov
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States.,A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| | - Matthew W Vetting
- Department of Biochemistry, Albert Einstein College of Medicine, New York, United States
| | - Nawar F Al-Obaidi
- Department of Biochemistry, Albert Einstein College of Medicine, New York, United States
| | - Henry Lin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Matthew J O'Meara
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - David A Scott
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - John H Morris
- Resource for Biocomputing, Visualization and Informatics, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Daniel Russel
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, New York, United States
| | - Andrei L Osterman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - John A Gerlt
- Institute for Genomic Biology, University of Illinois, Urbana, United States.,Department of Biochemistry, University of Illinois, Urbana, United States.,Department of Chemistry, University of Illinois, Urbana, United States
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
9
|
Kandoussi I, Lakhlili W, Taoufik J, Ibrahimi A. Docking analysis of verteporfin with YAP WW domain. Bioinformation 2017; 13:237-240. [PMID: 28943729 PMCID: PMC5602291 DOI: 10.6026/97320630013237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/07/2017] [Accepted: 07/08/2017] [Indexed: 11/23/2022] Open
Abstract
The YAP oncogene is a known cancer target. Therefore, it is of interest to understand the molecular docking interaction of verteporfin (a derivative of benzo-porphyrin) with the WW domain of YAP (clinically used for photo-dynamic therapy in macular degeneration) as a potential WW domain-ligand modulator by inhibition. A homology protein SWISS MODEL of the human YAP protein was constructed to dock (using AutoDock vina) with the PubChem verteporfin structure for interaction analysis. The docking result shows the possibilities of verteporfin interaction with the oncogenic transcription cofactor YAP having WW1 and WW2 domains. Thus, the ability of verteporfin to bind with the YAP WW domain having modulator activity is implied in this analysis.
Collapse
Affiliation(s)
- Ilham Kandoussi
- Laboratoire de Biotechnologie (MedBiotech), Faculté de Médecine et de Pharmacie de Rabat, Université Mohammed V Rabat, Morocco
| | - Wiame Lakhlili
- Laboratoire de Biotechnologie (MedBiotech), Faculté de Médecine et de Pharmacie de Rabat, Université Mohammed V Rabat, Morocco
| | - Jamal Taoufik
- Laboratoire de Chimie thérapeutique Faculté de Médecine et de Pharmacie de Rabat, Université Mohammed V Rabat,Morocco
| | - Azeddine Ibrahimi
- Laboratoire de Biotechnologie (MedBiotech), Faculté de Médecine et de Pharmacie de Rabat, Université Mohammed V Rabat, Morocco
| |
Collapse
|
10
|
Saenz-Méndez P, Eriksson M, Eriksson LA. Ligand Selectivity between the ADP-Ribosylating Toxins: An Inverse-Docking Study for Multitarget Drug Discovery. ACS OMEGA 2017; 2:1710-1719. [PMID: 30023642 PMCID: PMC6044789 DOI: 10.1021/acsomega.7b00010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/17/2017] [Indexed: 06/02/2023]
Abstract
Bacterial adenosine 5'-diphosphate-ribosylating toxins are encoded by several human pathogens, such as Pseudomonas aeruginosa (exotoxin A (ETA)), Corynebacterium diphtheriae (diphtheria toxin (DT)), and Vibrio cholerae (cholix toxin (CT)). The toxins modify eukaryotic elongation factor 2, an essential human enzyme in protein synthesis, thereby causing cell death. Targeting external virulence factors, such as the above toxins, is a promising alternative for developing new antibiotics, while at the same time avoiding drug resistance. This study aims to establish a reliable computational methodology to find a "silver bullet" able to target all three toxins. Herein, we have undertaken a detailed analysis of the active sites of ETA, DT, and CT, followed by the determination of the most appropriate selection of the size of the docking sphere. Thereafter, we tested two different approaches for normalizing the docking scores and used these to verify the best target (toxin) for each ligand. The results indicate that the methodology is suitable for identifying selective as well as multitoxin inhibitors, further validating the robustness of inverse docking for target-fishing experiments.
Collapse
Affiliation(s)
- Patricia Saenz-Méndez
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Göteborg, Sweden
- Computational
Chemistry and Biology Group, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Martin Eriksson
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Göteborg, Sweden
| | - Leif A. Eriksson
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Göteborg, Sweden
| |
Collapse
|
11
|
Targeting the Hippo Signaling Pathway for Tissue Regeneration and Cancer Therapy. Genes (Basel) 2016; 7:genes7090055. [PMID: 27589805 PMCID: PMC5042386 DOI: 10.3390/genes7090055] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
The Hippo signaling pathway is a highly-conserved developmental pathway that plays an essential role in organ size control, tumor suppression, tissue regeneration and stem cell self-renewal. The YES-associated protein (YAP) and the transcriptional co-activator with PDZ-binding motif (TAZ) are two important transcriptional co-activators that are negatively regulated by the Hippo signaling pathway. By binding to transcription factors, especially the TEA domain transcription factors (TEADs), YAP and TAZ induce the expression of growth-promoting genes, which can promote organ regeneration after injury. Therefore, controlled activation of YAP and TAZ can be useful for regenerative medicine. However, aberrant activation of YAP and TAZ due to deregulation of the Hippo pathway or overexpression of YAP/TAZ and TEADs can promote cancer development. Hence, pharmacological inhibition of YAP and TAZ may be a useful approach to treat tumors with high YAP and/or TAZ activity. In this review, we present the mechanisms regulating the Hippo pathway, the role of the Hippo pathway in tissue repair and cancer, as well as a detailed analysis of the different strategies to target the Hippo signaling pathway and the genes regulated by YAP and TAZ for regenerative medicine and cancer therapy.
Collapse
|
12
|
Lemos A, Leão M, Soares J, Palmeira A, Pinto M, Saraiva L, Sousa ME. Medicinal Chemistry Strategies to Disrupt the p53-MDM2/MDMX Interaction. Med Res Rev 2016; 36:789-844. [PMID: 27302609 DOI: 10.1002/med.21393] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/16/2016] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
The growth inhibitory activity of p53 tumor suppressor is tightly regulated by interaction with two negative regulatory proteins, murine double minute 2 (MDM2) and X (MDMX), which are overexpressed in about half of all human tumors. The elucidation of crystallographic structures of MDM2/MDMX complexes with p53 has been pivotal for the identification of several classes of inhibitors of the p53-MDM2/MDMX interaction. The present review provides in silico strategies and screening approaches used in drug discovery as well as an overview of the most relevant classes of small-molecule inhibitors of the p53-MDM2/MDMX interaction, their progress in pipeline, and highlights particularities of each class of inhibitors. Most of the progress made with high-throughput screening has led to the development of inhibitors belonging to the cis-imidazoline, piperidinone, and spiro-oxindole series. However, novel potent and selective classes of inhibitors of the p53-MDM2 interaction with promising antitumor activity are emerging. Even with the discovery of the 3D structure of complex p53-MDMX, only two small molecules were reported as selective p53-MDMX antagonists, WK298 and SJ-172550. Dual inhibition of the p53-MDM2/MDMX interaction has shown to be an alternative approach since it results in full activation of the p53-dependent pathway. The knowledge of structural requirements crucial to the development of small-molecule inhibitors of the p53-MDMs interactions has enabled the identification of novel antitumor agents with improved in vivo efficacy.
Collapse
Affiliation(s)
- Agostinho Lemos
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Mariana Leão
- UCIBIO/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Joana Soares
- UCIBIO/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Andreia Palmeira
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua de Bragas, 289, 4050-123, Porto, Portugal
| | - Lucília Saraiva
- UCIBIO/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Maria Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua de Bragas, 289, 4050-123, Porto, Portugal
| |
Collapse
|
13
|
The Roles of Mitochondrial Cation Channels Under Physiological Conditions and in Cancer. Handb Exp Pharmacol 2016; 240:47-69. [PMID: 27995386 DOI: 10.1007/164_2016_92] [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: 12/14/2022]
Abstract
Bioenergetics has become central to our understanding of pathological mechanisms as well as the development of new therapeutic strategies and as a tool for gauging disease progression in neurodegeneration, diabetes, cancer, and cardiovascular disease. The view is emerging that inner mitochondrial membrane (IMM) cation channels have a profound effect on mitochondrial function and, consequently, on the metabolic state and survival of the whole cell. Since disruption of the sustained integrity of mitochondria is strongly linked to human disease, pharmacological intervention offers a new perspective concerning neurodegenerative and cardiovascular diseases as well as cancer. This review summarizes our current knowledge regarding IMM cation channels and their roles under physiological conditions as well as in cancer, with special emphasis on potassium channels and the mammalian mitochondrial calcium uniporter.
Collapse
|
14
|
Surfing the Protein-Protein Interaction Surface Using Docking Methods: Application to the Design of PPI Inhibitors. Molecules 2015; 20:11569-603. [PMID: 26111183 PMCID: PMC6272567 DOI: 10.3390/molecules200611569] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/02/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Blocking protein-protein interactions (PPI) using small molecules or peptides modulates biochemical pathways and has therapeutic significance. PPI inhibition for designing drug-like molecules is a new area that has been explored extensively during the last decade. Considering the number of available PPI inhibitor databases and the limited number of 3D structures available for proteins, docking and scoring methods play a major role in designing PPI inhibitors as well as stabilizers. Docking methods are used in the design of PPI inhibitors at several stages of finding a lead compound, including modeling the protein complex, screening for hot spots on the protein-protein interaction interface and screening small molecules or peptides that bind to the PPI interface. There are three major challenges to the use of docking on the relatively flat surfaces of PPI. In this review we will provide some examples of the use of docking in PPI inhibitor design as well as its limitations. The combination of experimental and docking methods with improved scoring function has thus far resulted in few success stories of PPI inhibitors for therapeutic purposes. Docking algorithms used for PPI are in the early stages, however, and as more data are available docking will become a highly promising area in the design of PPI inhibitors or stabilizers.
Collapse
|
15
|
Santucci M, Vignudelli T, Ferrari S, Mor M, Scalvini L, Bolognesi ML, Uliassi E, Costi MP. The Hippo Pathway and YAP/TAZ-TEAD Protein-Protein Interaction as Targets for Regenerative Medicine and Cancer Treatment. J Med Chem 2015; 58:4857-73. [PMID: 25719868 DOI: 10.1021/jm501615v] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Hippo pathway is an important organ size control signaling network and the major regulatory mechanism of cell-contact inhibition. Yes associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are its targets and terminal effectors: inhibition of the pathway promotes YAP/TAZ translocation to the nucleus, where they interact with transcriptional enhancer associate domain (TEAD) transcription factors and coactivate the expression of target genes, promoting cell proliferation. Defects in the pathway can result in overgrowth phenotypes due to deregulation of stem-cell proliferation and apoptosis; members of the pathway are directly involved in cancer development. The pharmacological regulation of the pathway might be useful in cancer prevention, treatment, and regenerative medicine applications; currently, a few compounds can selectively modulate the pathway. In this review, we present an overview of the Hippo pathway, the sequence and structural analysis of YAP/TAZ, the known pharmacological modulators of the pathway, especially those targeting YAP/TAZ-TEAD interaction.
Collapse
Affiliation(s)
- Matteo Santucci
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Tatiana Vignudelli
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Stefania Ferrari
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Marco Mor
- ‡Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Laura Scalvini
- ‡Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Maria Laura Bolognesi
- §Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Elisa Uliassi
- §Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Paola Costi
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| |
Collapse
|
16
|
Schmidt D, Kolb P. Large-scale docking approaches to the kinome. J Cheminform 2014. [PMCID: PMC3980119 DOI: 10.1186/1758-2946-6-s1-p31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
17
|
Schuchardt BJ, Bhat V, Mikles DC, McDonald CB, Sudol M, Farooq A. Molecular basis of the binding of YAP transcriptional regulator to the ErbB4 receptor tyrosine kinase. Biochimie 2014; 101:192-202. [PMID: 24472438 DOI: 10.1016/j.biochi.2014.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/15/2014] [Indexed: 11/19/2022]
Abstract
The newly discovered transactivation function of ErbB4 receptor tyrosine kinase is believed to be mediated by virtue of the ability of its proteolytically-cleaved intracellular domain (ICD) to physically associate with YAP2 transcriptional regulator. In an effort to unearth the molecular basis of YAP2-ErbB4 interaction, we have conducted a detailed biophysical analysis of the binding of WW domains of YAP2 to PPXY motifs located within the ICD of ErbB4. Our data show that the WW1 domain of YAP2 binds to PPXY motifs within the ICD in a differential manner and that this behavior is by and large replicated by the WW2 domain. Remarkably, while both WW domains absolutely require the integrity of the PPXY consensus sequence, non-consensus residues within and flanking this motif do not appear to be critical for binding. In spite of this shared mode of binding, the WW domains of YAP2 display distinct conformational dynamics in complex with PPXY motifs derived from ErbB4. Collectively, our study lends new insights into the molecular basis of a key protein-protein interaction involved in a diverse array of cellular processes.
Collapse
Affiliation(s)
- Brett J Schuchardt
- Department of Biochemistry & Molecular Biology, Leonard Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Vikas Bhat
- Department of Biochemistry & Molecular Biology, Leonard Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David C Mikles
- Department of Biochemistry & Molecular Biology, Leonard Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Caleb B McDonald
- Department of Biochemistry & Molecular Biology, Leonard Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Marius Sudol
- Weis Center for Research, Geisinger Clinic, Danville, PA 17822, USA; Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Amjad Farooq
- Department of Biochemistry & Molecular Biology, Leonard Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
18
|
Patil SP. FOLICation: engineering approved drugs as potential p53-MDM2 interaction inhibitors for cancer therapy. Med Hypotheses 2013; 81:1104-7. [PMID: 24210632 DOI: 10.1016/j.mehy.2013.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/01/2013] [Accepted: 10/10/2013] [Indexed: 01/10/2023]
Abstract
Cancer is characterized by uncontrolled growth of abnormal cells leading to the formation of tumors. Normally, the pro-apoptotic p53 protein plays a central role in protecting cells against carcinogenesis. In almost 50% human tumor cells, however, the p53 protein is dysregulated by direct interaction with its negative regulator, the Murine Double Minute 2 (MDM2) protein. Therefore, blocking the p53-binding pocket on MDM2, leading to the activation of tumor suppressor p53 presents a novel therapeutic strategy against several types of cancers. The published crystal structure of MDM2 bound with the p53 binding domain has revealed that three key hydrophobic residues of p53 are buried deep into the binding cavity of MDM2 and thus are central to p53-MDM2 binding. Accordingly, several low-molecular-weight compounds have been developed that mimic these three hydrophobic residues and thus bind to the MDM2 pocket, leading in turn to inhibition of the deleterious p53-MDM2 interaction. It is noteworthy that these inhibitors also possess an additional hydrophilic group that is shown to be necessary as a "cover" protecting the hydrophobic interaction surface between inhibitor and MDM2 from surrounding solvent. In comparison, several FDA-approved drugs possess the three key hydrophobic features necessary for binding to MDM2, but lack the fourth hydrophilic moiety, thus possibly hindering their ability as potential p53-MDM2 interaction inhibitors. Therefore, we hypothesize that conjugation of hydrophilic vitamin folic acid or its analogs to these drugs (termed "FOLICation") may provide them with the much-needed hydrophilic cover and make them suitable for investigation as potentially novel p53-MDM2 inhibitors. We also anticipate that FOLICation of these drugs may further lead to their enhanced and selective uptake by cancer cells, owing to the significantly higher expression of folic acid receptors on cancer cells compared to normal cells.
Collapse
Affiliation(s)
- Sachin P Patil
- NanoBio Laboratory, Department of Chemical Engineering, Widener University, Chester, PA 19013, United States.
| |
Collapse
|
19
|
Finding protein targets for small biologically relevant ligands across fold space using inverse ligand binding predictions. Structure 2013; 20:1815-22. [PMID: 23141694 DOI: 10.1016/j.str.2012.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/14/2012] [Accepted: 09/16/2012] [Indexed: 01/12/2023]
Abstract
Inverse ligand binding prediction utilizes a few protein-ligand (drug) complexes to predict other secondary therapeutic and off-targets of a given drug molecule on a proteomic scale. We adapt two binding site predictors, FINDSITE and SMAP, to perform the inverse predictions and evaluate them on over 30 representative ligands. Use of just one complex allows the identification of other protein targets; the availability of additional complexes improves the results. Both methods offer comparable quality when using three complexes with diverse proteins. SMAP is better when fewer complexes are available, while FINDSITE provides stronger predictions for smaller ligands. We propose a consensus that combines (and outperforms) the two complementary approaches implemented by FINDSITE and SMAP. Most importantly, we demonstrate that these methods successfully find distant targets that belong to structurally different folds compared to the proteins in the input complexes.
Collapse
|
20
|
Structures of YAP protein domains reveal promising targets for development of new cancer drugs. Semin Cell Dev Biol 2012; 23:827-33. [PMID: 22609812 DOI: 10.1016/j.semcdb.2012.05.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/09/2012] [Accepted: 05/11/2012] [Indexed: 01/03/2023]
Abstract
YAP (Yes-associated protein) is a potent oncogene and a major effector of the mammalian Hippo tumor suppressor pathway. In this review, our emphasis is on the structural basis of how YAP recognizes its various cellular partners. In particular, we discuss the role of LATS kinase and AMOTL1 junction protein, two key cellular partners of YAP that bind to its WW domain, in mediating cytoplasmic localization of YAP and thereby playing a key role in the regulation of its transcriptional activity. Importantly, the crystal structure of an amino-terminal domain of YAP in complex with the carboxy-terminal domain of TEAD transcription factor was only recently solved at atomic resolution, while the structure of WW domain of YAP in complex with a peptide containing the PPxY motif has been available for more than a decade. We discuss how such structural information may be exploited for the rational development of novel anti-cancer therapeutics harboring greater efficacy coupled with low toxicity. We also embark on a brief discussion of how recent in silico studies led to identification of the cardiac glycoside digitoxin as a potential modulator of WW domain-ligand interactions. Conversely, dobutamine was identified in a screen of known drugs as a compound that promotes cytoplasmic localization of YAP, thereby resulting in growth suppressing activity. Finally, we discuss how a recent study on the dynamics of WW domain folding on a biologically critical time scale may provide a tool to generate repertoires of WW domain variants for regulation of the Hippo pathway toward desired, non-oncogenic outputs.
Collapse
|
21
|
Wilson CGM, Arkin MR. Small-molecule inhibitors of IL-2/IL-2R: lessons learned and applied. Curr Top Microbiol Immunol 2011; 348:25-59. [PMID: 20703966 DOI: 10.1007/82_2010_93] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The IL-2:IL-2R protein-protein interaction is of central importance to both healthy and diseased immune responses, and is one of the earliest examples of successful small-molecule inhibitor discovery against this target class. Drug-like inhibitors of IL-2 have been identified through a combination of fragment discovery, structure-based design, and medicinal chemistry; this discovery approach illustrates the importance of using a diverse range of complementary screening methods and analytical tools to achieve a comprehensive understanding of molecular recognition. The IL-2 story also provides insight into the dynamic nature of protein-protein interaction surfaces, their potential druggability, and the physical and chemical properties of effective small-molecule ligands. These lessons, from IL-2 and similar discovery programs, underscore an increasing awareness of the principles governing the development of drugs for protein-protein interactions.
Collapse
Affiliation(s)
- C G M Wilson
- Small Molecule Discovery Center, University of California, San Francisco, CA 94158, USA
| | | |
Collapse
|
22
|
Davis FP. Proteome-wide prediction of overlapping small molecule and protein binding sites using structure. MOLECULAR BIOSYSTEMS 2010; 7:545-57. [PMID: 21103609 DOI: 10.1039/c0mb00200c] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Small molecules that modulate protein-protein interactions are of great interest for chemical biology and therapeutics. Here I present a structure-based approach to predict 'bi-functional' sites able to bind both small molecule ligands and proteins, in proteins of unknown structure. First, I develop a homology-based annotation method that transfers binding sites of known three-dimensional structure onto protein sequences, predicting residues in ligand and protein binding sites with estimated true positive rates of 98% and 88%, respectively, at 1% false positive rates. Applying this method to the human proteome predicts 8463 proteins with bi-functional residues and correctly recovers the targets of known interaction modulators. Proteins with significantly (p < 0.01) more bi-functional residues than expected were found to be enriched in regulatory and depleted in metabolism functions. Finally, I demonstrate the utility of the method by describing examples of predicted overlap and evidence of their biological and therapeutic relevance. The results suggest that combining the structures of known binding sites with established fold detection algorithms can predict regions of protein-protein interfaces that are amenable to small molecule modulation. Open-source software and the results for several complete proteomes are available at http://pibase.janelia.org/homolobind.
Collapse
Affiliation(s)
- Fred P Davis
- Howard Hughes Medical Institute, Janelia Farm Research Campus, 19700 Helix Dr, Ashburn, VA 20147, USA.
| |
Collapse
|
23
|
Sigalov AB. The SCHOOL of nature: III. From mechanistic understanding to novel therapies. SELF/NONSELF 2010; 1:192-224. [PMID: 21487477 PMCID: PMC3047783 DOI: 10.4161/self.1.3.12794] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 06/10/2010] [Accepted: 06/11/2010] [Indexed: 11/19/2022]
Abstract
Protein-protein interactions play a central role in biological processes and thus represent an appealing target for innovative drug design and development. They can be targeted by small molecule inhibitors, modulatory peptides and peptidomimetics, which represent a superior alternative to protein therapeutics that carry many disadvantages. Considering that transmembrane signal transduction is an attractive process to therapeutically control multiple diseases, it is fundamentally and clinically important to mechanistically understand how signal transduction occurs. Uncovering specific protein-protein interactions critical for signal transduction, a general platform for receptor-mediated signaling, the signaling chain homooligomerization (SCHOOL) platform, suggests these interactions as universal therapeutic targets. Within the platform, the general principles of signaling are similar for a variety of functionally unrelated receptors. This suggests that global therapeutic strategies targeting key protein-protein interactions involved in receptor triggering and transmembrane signal transduction may be used to treat a diverse set of diseases. This also assumes that clinical knowledge and therapeutic strategies can be transferred between seemingly disparate disorders, such as T cell-mediated skin diseases and platelet disorders or combined to develop novel pharmacological approaches. Intriguingly, human viruses use the SCHOOL-like strategies to modulate and/or escape the host immune response. These viral mechanisms are highly optimized over the millennia, and the lessons learned from viral pathogenesis can be used practically for rational drug design. Proof of the SCHOOL concept in the development of novel therapies for atopic dermatitis, rheumatoid arthritis, cancer, platelet disorders and other multiple indications with unmet needs opens new horizons in therapeutics.
Collapse
|