101
|
Anoopkumar-Dukie S, Conere T, Sisk GD, Allshire A. Mitochondrial modulation of oxygen-dependent radiosensitivity in some human tumour cell lines. Br J Radiol 2009; 82:847-54. [PMID: 19366737 DOI: 10.1259/bjr/35746067] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oxygen-dependent radiosensitivity of tumour cells reflects direct oxidative damage to DNA, but non-nuclear mechanisms including signalling pathways may also contribute. Mitochondria are likely candidates because not only do they integrate signals from each of the main kinase pathways but mitochondrial kinases responsive to oxidative stress communicate to the rest of the cell. Using pharmacological and immunochemical methods, we tested the role of mitochondrial permeability transition (MPT) and the Bcl-2 proteins in oxygen-dependent radiosensitivity. Drug-treated or untreated cervical cancer HeLa, breast cancer MCF-7 and melanoma MeWo cell lines were irradiated at 6.2 Gy under normoxic and hypoxic conditions then allowed to proliferate for 7 days. The MPT blocker cyclosporin A (2 microM) strongly protected HeLa but not the other two lines against oxygen-dependent radiosensitivity. By contrast, bongkrekic acid (50 microM), which blocks MPT by targeting the adenine nucleotide transporter, had only marginal effect and calcineurin inhibitor FK-506 (0.1 microM) had none. Nor was evidence found for the modulation of oxygen-dependent radiosensitivity by Bax/Bcl-2 signalling, mitochondrial ATP-dependent potassium (mitoK(ATP)) channels or mitochondrial Ca(2+) uptake. In conclusion, calcineurin-independent protection by cyclosporin A suggests that MPT but not mitoK(ATP) or the mitochondrial apoptosis pathway plays a causal role in oxygen-dependent radiosensitivity of HeLa cells. Targeting MPT may therefore improve the effectiveness of radiotherapy in some solid tumours.
Collapse
Affiliation(s)
- S Anoopkumar-Dukie
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
102
|
Skolnick J, Brylinski M. FINDSITE: a combined evolution/structure-based approach to protein function prediction. Brief Bioinform 2009; 10:378-91. [PMID: 19324930 DOI: 10.1093/bib/bbp017] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A key challenge of the post-genomic era is the identification of the function(s) of all the molecules in a given organism. Here, we review the status of sequence and structure-based approaches to protein function inference and ligand screening that can provide functional insights for a significant fraction of the approximately 50% of ORFs of unassigned function in an average proteome. We then describe FINDSITE, a recently developed algorithm for ligand binding site prediction, ligand screening and molecular function prediction, which is based on binding site conservation across evolutionary distant proteins identified by threading. Importantly, FINDSITE gives comparable results when high-resolution experimental structures as well as predicted protein models are used.
Collapse
Affiliation(s)
- Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology 250 14th St NW, Atlanta, GA 30318, USA.
| | | |
Collapse
|
103
|
Uo T, Veenstra TD, Morrison RS. Histone deacetylase inhibitors prevent p53-dependent and p53-independent Bax-mediated neuronal apoptosis through two distinct mechanisms. J Neurosci 2009; 29:2824-32. [PMID: 19261878 PMCID: PMC2673506 DOI: 10.1523/jneurosci.6186-08.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 01/29/2009] [Accepted: 01/30/2009] [Indexed: 01/06/2023] Open
Abstract
Pharmacological manipulation of protein acetylation levels by histone deacetylase (HDAC) inhibitors represents a novel therapeutic strategy to treat neurodegeneration as well as cancer. However, the molecular mechanisms that determine how HDAC inhibition exerts a protective effect in neurons as opposed to a cytotoxic action in tumor cells has not been elucidated. We addressed this issue in cultured postnatal mouse cortical neurons whose p53-dependent and p53-independent intrinsic apoptotic programs require the proapoptotic multidomain protein, Bax. Despite promoting nuclear p53 accumulation, Class I/II HDAC inhibitors (HDACIs) protected neurons from p53-dependent cell death induced by camptothecin, etoposide, heterologous p53 expression or the MDM2 inhibitor, nutlin-3a. HDACIs suppressed p53-dependent PUMA expression, a critical signaling intermediate linking p53 to Bax activation, thus preventing postmitochondrial events including cleavage of caspase-9 and caspase-3. In human SH-SY5Y neuroblastoma cells, however, HDACIs were not able to prevent p53-dependent cell death. Moreover, HDACIs also prevented caspase-3 cleavage in postnatal cortical neurons treated with staurosporine, 3-nitropropionic acid and a Bcl-2 inhibitor, all of which require the presence of Bax but not p53 to promote apoptosis. Although these three toxic agents displayed a requirement for Bax, they did not promote PUMA induction. These results demonstrate that HDACIs block Bax-dependent cell death by two distinct mechanisms to prevent neuronal apoptosis, thus identifying for the first time a defined molecular target for their neuroprotective actions.
Collapse
Affiliation(s)
- Takuma Uo
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98195-6470, and
| | - Timothy D. Veenstra
- Laboratory of Proteomics and Analytical Technologies, Science Applications International Corporation-Frederick, Inc., National Cancer Institute at Frederick, Frederick, Maryland 21702-1201
| | - Richard S. Morrison
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98195-6470, and
| |
Collapse
|
104
|
Arisan ED, Kutuk O, Tezil T, Bodur C, Telci D, Basaga H. Small inhibitor of Bcl-2, HA14-1, selectively enhanced the apoptotic effect of cisplatin by modulating Bcl-2 family members in MDA-MB-231 breast cancer cells. Breast Cancer Res Treat 2009; 119:271-81. [DOI: 10.1007/s10549-009-0343-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 02/06/2009] [Indexed: 10/21/2022]
|
105
|
ApoG2 inhibits antiapoptotic Bcl-2 family proteins and induces mitochondria-dependent apoptosis in human lymphoma U937 cells. Anticancer Drugs 2009; 19:967-74. [PMID: 18827561 DOI: 10.1097/cad.0b013e32831087e8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lymphoma is one of the most common types of hematological malignancies and proteins from the Bcl-2 family are highly expressed in human lymphomas. Apogossypolone (ApoG2), the most potent gossypol derivative, has been classified as a novel small-molecule inhibitor of antiapoptotic Bcl-2 family proteins. Here, we assessed the in-vitro cytotoxicity of ApoG2 on human U937 lymphoma cells, and explored the underlying intracellular molecular mechanisms of ApoG2. Using the WST-8 assay, we found that ApoG2 inhibited growth of U937 cells in a dose-dependent and time-dependent manner, and the IC50 values were 30.08, 14.81, and 9.26 mumol/l for 24, 48, and 72 h treatments, respectively. ApoG2 also induced apoptosis in U937 cells, as noted through changes in morphological characteristics, including cellular internucleosomal DNA fragmentation and the appearance of a sub-G1 apoptotic peak. Treatment with ApoG2 downregulated Bcl-xL and Mcl-1 protein expression and blocked the binding of Bcl-2 with Bax protein. Furthermore, ApoG2 led to an abundant release of cytochrome c from mitochondria and a five-fold increase in the activity of caspase-3 and caspase-9. Taken together, our results suggest that ApoG2 could effectively suppress the growth of human lymphoma cell line U937 through the inhibition of the antiapoptotic Bcl-2 family proteins and the induction of mitochondria-dependent apoptotic cell death.
Collapse
|
106
|
Levesque MJ, Ichikawa K, Date S, Haga JH. Design of a grid service-based platform for in silico protein-ligand screenings. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2009; 93:73-82. [PMID: 18771812 PMCID: PMC2665129 DOI: 10.1016/j.cmpb.2008.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 06/13/2008] [Accepted: 07/11/2008] [Indexed: 05/26/2023]
Abstract
Grid computing offers the powerful alternative of sharing resources on a worldwide scale, across different institutions to run computationally intensive, scientific applications without the need for a centralized supercomputer. Much effort has been put into development of software that deploys legacy applications on a grid-based infrastructure and efficiently uses available resources. One field that can benefit greatly from the use of grid resources is that of drug discovery since molecular docking simulations are an integral part of the discovery process. In this paper, we present a scalable, reusable platform to choreograph large virtual screening experiments over a computational grid using the molecular docking simulation software DOCK. Software components are applied on multiple levels to create automated workflows consisting of input data delivery, job scheduling, status query, and collection of output to be displayed in a manageable fashion for further analysis. This was achieved using Opal OP to wrap the DOCK application as a grid service and PERL for data manipulation purposes, alleviating the requirement for extensive knowledge of grid infrastructure. With the platform in place, a screening of the ZINC 2,066,906 compound "drug-like" subset database against an enzyme's catalytic site was successfully performed using the MPI version of DOCK 5.4 on the PRAGMA grid testbed. The screening required 11.56 days laboratory time and utilized 200 processors over 7 clusters.
Collapse
Affiliation(s)
- Marshall J. Levesque
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093-0435
| | - Kohei Ichikawa
- Research Center of Socionetwork Strategies, The Institution of Economic and Political Studies, Kansai University, 3-3-35 Yamate-cho, Suita, Osaka, 564-8680 Japan
| | - Susumu Date
- Cybermedia Center, Osaka University, 5-1 Mihogaoka Ibaraki, Osaka 567-0047, Japan
| | - Jason H. Haga
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093-0435
| |
Collapse
|
107
|
Synthesis and anticancer activity in vitro of isothiochromeno[3,4-d]thiazole derivatives. ACTA ACUST UNITED AC 2008. [DOI: 10.2478/v10080-008-0044-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
108
|
Lessene G, Czabotar PE, Colman PM. BCL-2 family antagonists for cancer therapy. Nat Rev Drug Discov 2008; 7:989-1000. [DOI: 10.1038/nrd2658] [Citation(s) in RCA: 484] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
109
|
Abstract
BH3-only BCL-2 family proteins are effectors of canonical mitochondrial apoptosis. They discharge their pro-apoptotic functions through BH1-3 pro-apoptotic proteins such as BAX and BAK, while their activity is suppressed by BH1-4 anti-apoptotic BCL-2 family members. The precise mechanism by which BH3-only proteins mediate apoptosis remains unresolved. The existing data are consistent with three mutually non-exclusive models (1) displacement of BH1-3 proteins from complexes with BH1-4 proteins; (2) direct interaction with and conformational activation of BH1-3 proteins; and (3) membrane insertion and membrane remodeling. The BH3-only proteins appear to play critical roles in restraining cancer and inflammatory diseases such as rheumatoid arthritis. Molecules that mimic the effect of BH3-only proteins are being used in treatments against these diseases. The cell death activity of a subclass of BH3-only members (BNIP3 and BNIP3L) is linked to cardiomyocyte loss during heart failure. In addition to their established role in apoptosis, several BH3-only members also regulate diverse cellular functions in cell-cycle regulation, DNA repair and metabolism. Several members are implicated in the induction of autophagy and autophagic cell death, possibly through unleashing of the BH3-only autophagic effector Beclin 1 from complexes with BCL-2/BCL-xL. The Chapters included in the current Oncogene Review issues provide in-depth discussions on various aspects of major BH3-only proteins.
Collapse
Affiliation(s)
| | - G. Chinnadurai
- Institute for Molecular Virology, Saint Louis University School of Medicine, Doisy Research Center, 1100 South Grand Blvd, St. Louis, MO 63104
| |
Collapse
|
110
|
Abstract
BCL-2 was the first anti-death gene discovered, a milestone with far reaching implications for tumor biology. Multiple members of the human Bcl-2 family of apoptosis-regulating proteins have been identified, including six antiapoptotic, three structurally similar proapoptotic proteins and several structurally diverse proapoptotic interacting proteins that operate as upstream agonists or antagonists. These proteins, in turn, are regulated through myriad post-translational modifications and interactions with other proteins. Bcl-2-family proteins regulate all major types of cell death, including apoptosis, necrosis and autophagy, thus operating as nodal points at the convergence of multiple pathways with broad relevance to oncology. Experimental therapies targeting Bcl-2-family mRNAs or proteins are currently in clinical testing, raising hopes that a new class of anticancer drugs may soon be available.
Collapse
|
111
|
Lama D, Sankararamakrishnan R. Anti-apoptotic Bcl-XL protein in complex with BH3 peptides of pro-apoptotic Bak, Bad, and Bim proteins: comparative molecular dynamics simulations. Proteins 2008; 73:492-514. [PMID: 18452209 DOI: 10.1002/prot.22075] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The Bcl-2 family of proteins plays a central role in the regulation of mitochondrial outer-membrane permeabilization, a critical step in apoptosis. Heterodimerization between the pro- and anti-apoptotic members of Bcl-2 family is a key event in this process. Anti-apoptotic proteins have high levels of expression in many cancers and they have different affinities for different pro-apoptotic proteins. Experimentally determined structures of all members of Bcl-2 proteins have remarkably similar helical fold despite poor amino acid sequence identity. Peptides representing BH3 region of pro-apoptotic proteins have been shown to bind the hydrophobic cleft of anti-apoptotic proteins and this segment is responsible in modulating the apoptotic pathways in living cells. Understanding the molecular basis of protein-protein recognition is required to develop inhibitors specific to a particular anti-apoptotic protein. We have carried out molecular dynamics simulations on the anti-apoptotic Bcl-X(L) protein in complex with three different BH3 peptides derived from pro-apoptotic Bak, Bad and Bim proteins. Each complex structure was simulated for a period of 50 ns after 2.5 ns equilibration. Analysis of the simulation results showed that in the Bcl-X(L) protein, the helix containing the BH3 region is more flexible than other helices in all three simulations. A network of strong hydrophobic interactions exists between four of the six helices and they contribute significantly to the stability of this helix bundle protein. Analysis of Bcl-X(L)-BH3 peptide interactions reveals the role of loop residues in the protein-peptide interactions in all three simulations. Bad and Bim peptides maintain strong hydrophobic and hydrophilic interactions with the helix preceding the central hydrophobic helix. Residues from this helix interact with an Arg residue in Bad and Bim peptides. This Arg residue is next to the conserved Leu residue and is replaced by Ala in Bak. Absence of these interactions and the helix propensity are likely to be the cause for Bak peptide's weaker binding affinity with the Bcl-X(L) protein. The results of this study have implications in the design of Bcl-X(L)-specific inhibitors.
Collapse
Affiliation(s)
- Dilraj Lama
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | | |
Collapse
|
112
|
Bernardo PH, Wan KF, Sivaraman T, Xu J, Moore FK, Hung AW, Mok HYK, Yu VC, Chai CLL. Structure−Activity Relationship Studies of Phenanthridine-Based Bcl-XL Inhibitors. J Med Chem 2008; 51:6699-710. [DOI: 10.1021/jm8005433] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Paul H. Bernardo
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Kah-Fei Wan
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Thirunavukkarasu Sivaraman
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Jin Xu
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Felicity K. Moore
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Alvin W. Hung
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Henry Y. K. Mok
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Victor C. Yu
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| | - Christina L. L. Chai
- Institute of Chemical & Engineering Sciences, Agency for Science Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Institute of Molecular & Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Singapore 138673, Departmental of Biological Sciences, National University of Singapore, Singapore 117543
| |
Collapse
|
113
|
Sun Y, Wu J, Aboukameel A, Banerjee S, Arnold AA, Chen J, Nikolovska-Coleska Z, Lin Y, Ling X, Yang D, Wang S, Al-Katib A, Mohammad RM. Apogossypolone, a nonpeptidic small molecule inhibitor targeting Bcl-2 family proteins, effectively inhibits growth of diffuse large cell lymphoma cells in vitro and in vivo. Cancer Biol Ther 2008; 7:1418-26. [PMID: 18769131 DOI: 10.4161/cbt.7.9.6430] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Apogossypolone (ApoG2) is a semi-synthesized derivative of gossypol. The principal objective of this study was to compare stability and toxicity between ApoG2 and gossypol, and to evaluate anti-lymphoma activity of ApoG2 in vitro and in vivo. ApoG2 shows better stability when compared with a racemic gossypol and can be better tolerated by mice compared to gossypol. ApoG2 showed significant inhibition of cell proliferation of WSU-DLCL(2) and primary cells obtained from lymphoma patients, whereas it displayed no toxicity on normal peripheral blood lymphocytes. For a treatment of 72 h, the IC(50) of ApoG2 was determined to be 350 nM against WSU-DLCL2 cells. Treatment with ApoG2 at 600 mg/kg resulted in significant growth inhibition of WSU-DLCL(2) xenografts. When combined with CHOP, ApoG2 displayed even more complete inhibition of tumor growth. ApoG2 binds to purified recombinant Bcl-2, Mcl-1 and Bcl-X(L) proteins with high affinity and is shown to block the formation of heterodimers between Bcl-X(L) and Bim. For a treatment of 72 h, ApoG2 induced a maximum of 32% of apoptotic cell death. Western blot experiments showed that treatment with ApoG2 led to cleavage of caspase-3, caspase-9 and PARP. Moreover, pretreatment of DLCL(2) cells with caspase-3, -9 and broad spectrum caspase inhibitors significantly blocked growth inhibition induced by ApoG2. In conclusion, ApoG2 effectively inhibits growth of DLCL(2) cells at least partly by inducing apoptosis. It is an attractive small molecule inhibitor of the Bcl-2 family proteins to be developed further for the treatment of diffuse large cell lymphoma.
Collapse
Affiliation(s)
- Yuan Sun
- Division of Hematology and Oncology, Department of Internal Medicine, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Eswar N, Webb B, Marti-Renom MA, Madhusudhan MS, Eramian D, Shen MY, Pieper U, Sali A. Comparative protein structure modeling using MODELLER. ACTA ACUST UNITED AC 2008; Chapter 2:Unit 2.9. [PMID: 18429317 DOI: 10.1002/0471140864.ps0209s50] [Citation(s) in RCA: 761] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Functional characterization of a protein sequence is a common goal in biology, and is usually facilitated by having an accurate three-dimensional (3-D) structure of the studied protein. In the absence of an experimentally determined structure, comparative or homology modeling can sometimes provide a useful 3-D model for a protein that is related to at least one known protein structure. Comparative modeling predicts the 3-D structure of a given protein sequence (target) based primarily on its alignment to one or more proteins of known structure (templates). The prediction process consists of fold assignment, target-template alignment, model building, and model evaluation. This unit describes how to calculate comparative models using the program MODELLER and discusses all four steps of comparative modeling, frequently observed errors, and some applications. Modeling lactate dehydrogenase from Trichomonas vaginalis (TvLDH) is described as an example. The download and installation of the MODELLER software is also described.
Collapse
Affiliation(s)
- Narayanan Eswar
- University of California at San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Brylinski M, Skolnick J. Q-Dock: Low-resolution flexible ligand docking with pocket-specific threading restraints. J Comput Chem 2008; 29:1574-88. [PMID: 18293308 PMCID: PMC2726574 DOI: 10.1002/jcc.20917] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The rapidly growing number of theoretically predicted protein structures requires robust methods that can utilize low-quality receptor structures as targets for ligand docking. Typically, docking accuracy falls off dramatically when apo or modeled receptors are used in docking experiments. Low-resolution ligand docking techniques have been developed to deal with structural inaccuracies in predicted receptor models. In this spirit, we describe the development and optimization of a knowledge-based potential implemented in Q-Dock, a low-resolution flexible ligand docking approach. Self-docking experiments using crystal structures reveals satisfactory accuracy, comparable with all-atom docking. All-atom models reconstructed from Q-Dock's low-resolution models can be further refined by even a simple all-atom energy minimization. In decoy-docking against distorted receptor models with a root-mean-square deviation, RMSD, from native of approximately 3 A, Q-Dock recovers on average 15-20% more specific contacts and 25-35% more binding residues than all-atom methods. To further improve docking accuracy against low-quality protein models, we propose a pocket-specific protein-ligand interaction potential derived from weakly homologous threading holo-templates. The success rate of Q-Dock employing a pocket-specific potential is 6.3 times higher than that previously reported for the Dolores method, another low-resolution docking approach.
Collapse
Affiliation(s)
- Michal Brylinski
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250 14th Street NW, Atlanta, GA 30318, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250 14th Street NW, Atlanta, GA 30318, USA
| |
Collapse
|
116
|
[Value of targeted treatment for testicular cancer: from molecular approaches to clinical possibilities]. Urologe A 2008; 47:1328-33. [PMID: 18587552 DOI: 10.1007/s00120-008-1750-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Due to the introduction of tyrosine kinase-inhibitors in the treatment of metastatic renal cell cancer, targeted therapy raises hopes for other urological tumors as well. Even if excellent cure rates, achieved by standardization of diagnosis und therapy, have made testicular cancer a curable disease, up to 6% of young patients still die from tumors refractory to therapy. The quality of life of patients in advanced stages needing aggressive treatment should be improved by new therapies with reduced side effects. The role of tyrosine kinase inhibitors and angiogenesis inhibitors as well as intervention in the cell cycle and induction of apoptosis are discussed.
Collapse
|
117
|
Casey FP, Davey NE, Baran I, Varekova RS, Shields DC. Web Server To Identify Similarity of Amino Acid Motifs to Compounds (SAAMCO). J Chem Inf Model 2008; 48:1524-9. [DOI: 10.1021/ci8000474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Fergal P. Casey
- UCD Conway Institute of Biomolecular and Biomedical Sciences and Complex and Adaptive Systems Laboratory (CASL), University College Dublin, Dublin, Ireland, Siemens Research Ireland, Dublin, Ireland, and ANF Data, a Siemens Company, Brno, Czech Republic
| | - Norman E. Davey
- UCD Conway Institute of Biomolecular and Biomedical Sciences and Complex and Adaptive Systems Laboratory (CASL), University College Dublin, Dublin, Ireland, Siemens Research Ireland, Dublin, Ireland, and ANF Data, a Siemens Company, Brno, Czech Republic
| | - Ivan Baran
- UCD Conway Institute of Biomolecular and Biomedical Sciences and Complex and Adaptive Systems Laboratory (CASL), University College Dublin, Dublin, Ireland, Siemens Research Ireland, Dublin, Ireland, and ANF Data, a Siemens Company, Brno, Czech Republic
| | - Radka Svobodova Varekova
- UCD Conway Institute of Biomolecular and Biomedical Sciences and Complex and Adaptive Systems Laboratory (CASL), University College Dublin, Dublin, Ireland, Siemens Research Ireland, Dublin, Ireland, and ANF Data, a Siemens Company, Brno, Czech Republic
| | - Denis C. Shields
- UCD Conway Institute of Biomolecular and Biomedical Sciences and Complex and Adaptive Systems Laboratory (CASL), University College Dublin, Dublin, Ireland, Siemens Research Ireland, Dublin, Ireland, and ANF Data, a Siemens Company, Brno, Czech Republic
| |
Collapse
|
118
|
Ma XH, Wang R, Yang SY, Li ZR, Xue Y, Wei YC, Low BC, Chen YZ. Evaluation of virtual screening performance of support vector machines trained by sparsely distributed active compounds. J Chem Inf Model 2008; 48:1227-37. [PMID: 18533644 DOI: 10.1021/ci800022e] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Virtual screening performance of support vector machines (SVM) depends on the diversity of training active and inactive compounds. While diverse inactive compounds can be routinely generated, the number and diversity of known actives are typically low. We evaluated the performance of SVM trained by sparsely distributed actives in six MDDR biological target classes composed of a high number of known actives (983-1645) of high, intermediate, and low structural diversity (muscarinic M1 receptor agonists, NMDA receptor antagonists, thrombin inhibitors, HIV protease inhibitors, cephalosporins, and renin inhibitors). SVM trained by regularly sparse data sets of 100 actives show improved yields at substantially reduced false-hit rates compared to those of published studies and those of Tanimoto-based similarity searching method based on the same data sets and molecular descriptors. SVM trained by very sparse data sets of 40 actives (2.4%-4.1% of the known actives) predicted 17.5-39.5%, 23.0-48.1%, and 70.2-92.4% of the remaining 943-1605 actives in the high, intermediate, and low diversity classes, respectively, 13.8-68.7% of which are outside the training compound families. SVM predicted 99.97% and 97.1% of the 9.997 M PUBCHEM and 167K remaining MDDR compounds as inactive and 2.6%-8.3% of the 19,495-38,483 MDDR compounds similar to the known actives as active. These suggest that SVM has substantial capability in identifying novel active compounds from sparse active data sets at low false-hit rates.
Collapse
Affiliation(s)
- X H Ma
- Centre for Computational Science and Engineering, National University of Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
119
|
Boehm AL, Sen M, Seethala R, Gooding WE, Freilino M, Wong SMY, Wang S, Johnson DE, Grandis JR. Combined targeting of epidermal growth factor receptor, signal transducer and activator of transcription-3, and Bcl-X(L) enhances antitumor effects in squamous cell carcinoma of the head and neck. Mol Pharmacol 2008; 73:1632-42. [PMID: 18326051 PMCID: PMC3437602 DOI: 10.1124/mol.107.044636] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) is a leading cause of cancer deaths worldwide. Epidermal growth factor receptor (EGFR), an upstream mediator of signal transducer and activator of transcription (STAT)-3 is overexpressed in a variety of cancers, including SCCHN. Therapies such as monoclonal antibodies and tyrosine kinase inhibitors targeting EGFR have demonstrated limited antitumor efficacy, which may be explained, in part, by persistent STAT3 activation despite EGFR inhibition. STAT3 activation induces expression of target genes in SCCHN, including Bcl-X(L), a mediator of antiapoptotic activity. Bcl-X(L) is commonly overexpressed in SCCHN where it correlates with chemoresistance, making it a potential therapeutic target. Targeting the EGFR-STAT3-Bcl-X(L) pathway at several levels, including the upstream receptor, the intracellular transcription factor, and the downstream target gene, has not been investigated previously. Using erlotinib, an EGFR-specific reversible tyrosine kinase inhibitor in combination with a STAT3 transcription factor decoy, we found enhanced antitumor effects in vitro and in vivo. The combination of the STAT3 decoy and gossypol, a small molecule targeting Bcl-X(L), also yielded enhanced inhibition of cell proliferation. The triple combination of erlotinib, STAT3 decoy, and gossypol further enhanced cell growth inhibition and apoptosis in vitro, and it down-regulated signaling molecules further downstream of the EGFR-STAT3 signaling pathway, such as cyclin D1. These results suggest that combined targeting of several components of an oncogenic signaling pathway may be an effective therapeutic strategy for SCCHN.
Collapse
Affiliation(s)
- Amanda L Boehm
- Department of Pathology, University of Pittsburgh, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Labi V, Grespi F, Baumgartner F, Villunger A. Targeting the Bcl-2-regulated apoptosis pathway by BH3 mimetics: a breakthrough in anticancer therapy? Cell Death Differ 2008; 15:977-87. [PMID: 18369371 PMCID: PMC4563920 DOI: 10.1038/cdd.2008.37] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Induction of apoptosis in tumor cells by direct activation of the Bcl-2-regulated apoptosis pathway by small molecule drugs carries high hopes to overcome the shortcomings of current anticancer therapies. This novel therapy concept builds on emerging insights into how Bcl-2-like molecules maintain mitochondrial integrity and how pro-apoptotic BH3-only proteins lead to its disruption. Means to unleash the pro-apoptotic potential of BH3-only proteins in tumor cells, or to bypass the need for BH3-only proteins by directly blocking possible interactions of Bcl-2-like pro-survival molecules with Bax and/or Bak, constitute interesting options for the design of novel anticancer therapies. For the optimization and clinical implementation of these novel anticancer strategies, a detailed understanding of the role of individual BH3-only proteins in cell death signaling in healthy cells and during tumor suppression is required. In this review, we will touch on the latest findings on BH3-only protein function and attempts to define the molecular properties of the so-called 'BH3 mimetics,' a novel class of anticancer agents, able to prompt apoptosis in tumor cells, regardless of their p53 or Bcl-2 status.
Collapse
Affiliation(s)
- V Labi
- Division of Developmental Immunology, Department of Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - F Grespi
- Division of Developmental Immunology, Department of Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - F Baumgartner
- Division of Developmental Immunology, Department of Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - A Villunger
- Division of Developmental Immunology, Department of Biocenter, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
121
|
Dömling A, Antuch W, Beck B, Schauer-Vukasinović V. Isosteric exchange of the acylsulfonamide moiety in Abbott's Bcl-XL protein interaction antagonist. Bioorg Med Chem Lett 2008; 18:4115-7. [PMID: 18583128 DOI: 10.1016/j.bmcl.2008.05.096] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 05/21/2008] [Accepted: 05/22/2008] [Indexed: 10/22/2022]
Abstract
A multi-component reaction strategy was used for the fast and efficient synthesis of amide isosteres of known Bcl-2 inhibitors capable of disrupting protein-protein interactions. Ugi reaction and a subsequent nucleophilic aromatic substitution reaction provide a versatile path to libraries of compounds similar to Abbott's acylsulfonamides. Modeling arguments are used to explain the inferior activity of the amide as opposed to the sulfonamide series.
Collapse
Affiliation(s)
- Alexander Dömling
- Department of Pharmaceutical Sciences, University of Pittsburgh, PA 15261, USA.
| | | | | | | |
Collapse
|
122
|
Eswar N, Webb B, Marti-Renom MA, Madhusudhan MS, Eramian D, Shen MY, Pieper U, Sali A. Comparative protein structure modeling using Modeller. ACTA ACUST UNITED AC 2008; Chapter 5:Unit-5.6. [PMID: 18428767 DOI: 10.1002/0471250953.bi0506s15] [Citation(s) in RCA: 1821] [Impact Index Per Article: 107.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Functional characterization of a protein sequence is one of the most frequent problems in biology. This task is usually facilitated by accurate three-dimensional (3-D) structure of the studied protein. In the absence of an experimentally determined structure, comparative or homology modeling can sometimes provide a useful 3-D model for a protein that is related to at least one known protein structure. Comparative modeling predicts the 3-D structure of a given protein sequence (target) based primarily on its alignment to one or more proteins of known structure (templates). The prediction process consists of fold assignment, target-template alignment, model building, and model evaluation. This unit describes how to calculate comparative models using the program MODELLER and discusses all four steps of comparative modeling, frequently observed errors, and some applications. Modeling lactate dehydrogenase from Trichomonas vaginalis (TvLDH) is described as an example. The download and installation of the MODELLER software is also described.
Collapse
Affiliation(s)
- Narayanan Eswar
- University of California at San Francisco San Francisco, California
| | - Ben Webb
- University of California at San Francisco San Francisco, California
| | | | - M S Madhusudhan
- University of California at San Francisco San Francisco, California
| | - David Eramian
- University of California at San Francisco San Francisco, California
| | - Min-Yi Shen
- University of California at San Francisco San Francisco, California
| | - Ursula Pieper
- University of California at San Francisco San Francisco, California
| | - Andrej Sali
- University of California at San Francisco San Francisco, California
| |
Collapse
|
123
|
Radogna F, Cristofanon S, Paternoster L, D'Alessio M, De Nicola M, Cerella C, Dicato M, Diederich M, Ghibelli L. Melatonin antagonizes the intrinsic pathway of apoptosis via mitochondrial targeting of Bcl-2. J Pineal Res 2008; 44:316-25. [PMID: 18339127 DOI: 10.1111/j.1600-079x.2007.00532.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have recently shown that melatonin antagonizes damage-induced apoptosis by interaction with the MT-1/MT-2 plasma membrane receptors. Here, we show that melatonin interferes with the intrinsic pathway of apoptosis at the mitochondrial level. In response to an apoptogenic stimulus, melatonin allows mitochondrial translocation of the pro-apoptotic protein Bax, but it impairs its activation/dimerization The downstream apoptotic events, i.e. cytochrome c release, caspase 9 and 3 activation and nuclear vesiculation are equally impaired, indicating that melatonin interferes with Bax activation within mitochondria. Interestingly, we found that melatonin induces a strong re-localization of Bcl-2, the main Bax antagonist to mitochondria, suggesting that Bax activation may in fact be antagonized by Bcl-2 at the mitochondrial level. Indeed, we inhibit the melatonin anti-apoptotic effect (i) by silencing Bcl-2 with small interfering RNAs, or with small-molecular inhibitors targeted at the BH3 binding pocket in Bcl-2 (i.e. the one interacting with Bax); and (ii) by inhibiting melatonin-induced Bcl-2 mitochondrial re-localization with the MT1/MT2 receptor antagonist luzindole. This evidence provides a mechanism that may explain how melatonin through interaction with the MT1/MT2 receptors, elicits a pathway that interferes with the Bcl-2 family, thus modulating the cell life/death balance.
Collapse
Affiliation(s)
- Flavia Radogna
- Dipartimento di Biologia, Università di Roma Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Taylor P, Blackburn E, Sheng YG, Harding S, Hsin KY, Kan D, Shave S, Walkinshaw MD. Ligand discovery and virtual screening using the program LIDAEUS. Br J Pharmacol 2008; 153 Suppl 1:S55-67. [PMID: 18037921 PMCID: PMC2268042 DOI: 10.1038/sj.bjp.0707532] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2007] [Revised: 09/27/2007] [Accepted: 10/04/2007] [Indexed: 02/03/2023] Open
Abstract
This paper discusses advances in docking and scoring approaches with examples from the high-throughput virtual screening program LIDAEUS. We describe the discovery of small molecule inhibitors for the immunophilin CypA, the cyclin-dependent kinase CDK2 and the cyclapolin series of potent Polo-like kinase inhibitors. These results are discussed in the context of advances in massively parallel computing and in the development of annotated databases.
Collapse
Affiliation(s)
- P Taylor
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - E Blackburn
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - Y G Sheng
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - S Harding
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - K-Y Hsin
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - D Kan
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - S Shave
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| | - M D Walkinshaw
- The Centre for Translational and Chemical Biology, The University of Edinburgh, Michael Swann Building, King's Buildings, Mayfield Road, Edinburgh, UK
| |
Collapse
|
125
|
Murray JK, Sadowsky JD, Scalf M, Smith LM, Tomita Y, Gellman SH. Exploration of Structure−Activity Relationships among Foldamer Ligands for a Specific Protein Binding Site via Parallel and Split-and-Mix Library Synthesis. ACTA ACUST UNITED AC 2008; 10:204-15. [DOI: 10.1021/cc700153z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Justin K. Murray
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057
| | - Jack D. Sadowsky
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057
| | - Mark Scalf
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057
| | - Lloyd M. Smith
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057
| | - York Tomita
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057
| |
Collapse
|
126
|
Tian D, Das SGK, Doshi JM, Peng J, Lin J, Xing C. sHA 14-1, a stable and ROS-free antagonist against anti-apoptotic Bcl-2 proteins, bypasses drug resistances and synergizes cancer therapies in human leukemia cell. Cancer Lett 2008; 259:198-208. [PMID: 18037229 PMCID: PMC2693013 DOI: 10.1016/j.canlet.2007.10.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 10/08/2007] [Accepted: 10/10/2007] [Indexed: 11/23/2022]
Abstract
HA 14-1, a small-molecule antagonist against anti-apoptotic Bcl-2 proteins, was demonstrated to induce selective cytotoxicity toward malignant cells and to overcome drug resistance. Due to its poor stability and the reactive oxygen species (ROS) generated by its decomposition, chemical modification of HA 14-1 is needed for its future development. We have synthesized a stabilized analog of HA 14-1--sHA 14-1, which did not induce the formation of ROS. As expected from a putative antagonist against anti-apoptotic Bcl-2 proteins like HA 14-1, sHA 14-1 disrupted the binding interaction of a Bak BH3 peptide with Bcl-2 or Bcl-X(L) protein, inhibited the growth of tumor cells through the induction of apoptosis, and circumvented the drug resistance induced by the over-expression of anti-apoptotic Bcl-2 and Bcl-X(L) proteins. Interestingly, the impairment of extrinsic apoptotic pathway induced moderate resistance to sHA 14-1. The moderate resistance suggested that sHA 14-1 generated part of its apoptotic stress through the intrinsic pathway, possibly through its antagonism against anti-apoptotic Bcl-2 proteins. The resistance indicated that sHA 14-1 generated apoptotic stress through the extrinsic apoptotic pathway as well. The ability of sHA 14-1 to induce apoptotic stress through both pathways was further supported by the synergism of sHA 14-1 towards the cytotoxicities of Fas ligand and dexamethasone in Jurkat cells. Taken together, these findings suggest that sHA 14-1 may represent a promising candidate for the treatment of drug-resistant cancers either as a monotherapy or in combination with current cancer therapies.
Collapse
Affiliation(s)
- Defeng Tian
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis MN 55455
| | | | - Jignesh M. Doshi
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis MN 55455
| | - Jun Peng
- Department of Biochemistry and Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190
| | - Jialing Lin
- Department of Biochemistry and Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis MN 55455
| |
Collapse
|
127
|
Tang G, Nikolovska-Coleska Z, Qiu S, Yang CY, Guo J, Wang S. Acylpyrogallols as Inhibitors of Antiapoptotic Bcl-2 Proteins. J Med Chem 2008; 51:717-20. [DOI: 10.1021/jm701358v] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
128
|
Enyedy IJ, Egan WJ. Can we use docking and scoring for hit-to-lead optimization? J Comput Aided Mol Des 2008; 22:161-8. [PMID: 18183356 DOI: 10.1007/s10822-007-9165-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2007] [Accepted: 12/18/2007] [Indexed: 10/22/2022]
Abstract
Docking and scoring is currently one of the tools used for hit finding and hit-to-lead optimization when structural information about the target is known. Docking scores have been found useful for optimizing ligand binding to reproduce experimentally observed binding modes. The question is, can docking and scoring be used reliably for hit-to-lead optimization? To illustrate the challenges of scoring for hit-to-lead optimization, the relationship of docking scores with experimentally determined IC(50) values measured in-house were tested. The influences of the particular target, crystal structure, and the precision of the scoring function on the ability to differentiate between actives and inactives were analyzed by calculating the area under the curve of receiver operator characteristic curves for docking scores. It was found that for the test sets considered, MW and sometimes ClogP were as useful as GlideScores and no significant difference was observed between SP and XP scores for differentiating between actives and inactives. Interpretation by an expert is still required to successfully utilize docking and scoring in hit-to-lead optimization.
Collapse
Affiliation(s)
- Istvan J Enyedy
- Global Discovery Chemistry, Computer-Aided Drug Discovery, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | | |
Collapse
|
129
|
Apoptotic pathways in tumor progression and therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 615:47-79. [PMID: 18437891 DOI: 10.1007/978-1-4020-6554-5_4] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apoptosis is a cell suicide program that plays a critical role in development and tissue homeostasis. The ability of cancer cells to evade this programmed cell death (PCD) is a major characteristic that enables their uncontrolled growth. The efficiency of chemotherapy in killing such cells depends on the successful induction of apoptosis, since defects in apoptosis signaling are a major cause of drug resistance. Over the past decades, much progress has been made in our understanding of apoptotic signaling pathways and their dysregulation in cancer progression and therapy. These advances have provided new molecular targets for proapoptotic cancer therapies that have recently been used in drug development. While most of those therapies are still at the preclinical stage, some of them have shown much promise in the clinic. Here, we review our current knowledge of apoptosis regulation in cancer progression and therapy, as well as the new molecular targeted molecules that are being developed to reinstate cancer cell death.
Collapse
|
130
|
Del Gaizo Moore V, Letai A. Rational design of therapeutics targeting the BCL-2 family: are some cancer cells primed for death but waiting for a final push? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 615:159-75. [PMID: 18437895 DOI: 10.1007/978-1-4020-6554-5_8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
A mechanism for circumventing apoptosis prevalent in many cancer cells is the overexpression of antiapoptotic BCL-2 family members. Upregulated expression of BCL-2 may be required to permit ongoing death signaling without a cellular response. Therefore, antagonizing BCL-2 function may cause death in many cancer cells. The selection for expression of BCL-2 or other antiapoptotic proteins during oncogenesis may derive from these proteins' ability to bind and sequester proapoptotic BH3-only proteins. This situation may be advantageous from a therapeutic viewpoint because cancer cells may be distinguished from normal cells by being primed with death signals. There are several strategies currently under investigation that may lead to improved treatment of many cancers by taking advantage of these differences.
Collapse
Affiliation(s)
- Victoria Del Gaizo Moore
- Medical Oncology, Dana-Farber Cancer Institute, 44 Binney Street, Dana 530B, Boston, MA 02115, USA
| | | |
Collapse
|
131
|
Jiang X, Kong W, Chen J, Ma S. Intermolecular sequential [4 + 2]-cycloaddition–aromatization reaction of aryl-substituted allenes with DMAD affording phenanthrene and naphthalene derivatives. Org Biomol Chem 2008; 6:3606-10. [DOI: 10.1039/b808767a] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
132
|
Han LY, Ma XH, Lin HH, Jia J, Zhu F, Xue Y, Li ZR, Cao ZW, Ji ZL, Chen YZ. A support vector machines approach for virtual screening of active compounds of single and multiple mechanisms from large libraries at an improved hit-rate and enrichment factor. J Mol Graph Model 2007; 26:1276-86. [PMID: 18218332 DOI: 10.1016/j.jmgm.2007.12.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2007] [Revised: 12/05/2007] [Accepted: 12/05/2007] [Indexed: 01/04/2023]
Abstract
Support vector machines (SVM) and other machine-learning (ML) methods have been explored as ligand-based virtual screening (VS) tools for facilitating lead discovery. While exhibiting good hit selection performance, in screening large compound libraries, these methods tend to produce lower hit-rate than those of the best performing VS tools, partly because their training-sets contain limited spectrum of inactive compounds. We tested whether the performance of SVM can be improved by using training-sets of diverse inactive compounds. In retrospective database screening of active compounds of single mechanism (HIV protease inhibitors, DHFR inhibitors, dopamine antagonists) and multiple mechanisms (CNS active agents) from large libraries of 2.986 million compounds, the yields, hit-rates, and enrichment factors of our SVM models are 52.4-78.0%, 4.7-73.8%, and 214-10,543, respectively, compared to those of 62-95%, 0.65-35%, and 20-1200 by structure-based VS and 55-81%, 0.2-0.7%, and 110-795 by other ligand-based VS tools in screening libraries of >or=1 million compounds. The hit-rates are comparable and the enrichment factors are substantially better than the best results of other VS tools. 24.3-87.6% of the predicted hits are outside the known hit families. SVM appears to be potentially useful for facilitating lead discovery in VS of large compound libraries.
Collapse
Affiliation(s)
- L Y Han
- Bioinformatics and Drug Design Group, Department of Pharmacy, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore 117543, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Zhang L, Ming L, Yu J. BH3 mimetics to improve cancer therapy; mechanisms and examples. Drug Resist Updat 2007; 10:207-17. [PMID: 17921043 PMCID: PMC2265791 DOI: 10.1016/j.drup.2007.08.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 08/27/2007] [Accepted: 08/29/2007] [Indexed: 11/22/2022]
Abstract
Tumor cell survival is highly dependent on the expression of certain pro-survival Bcl-2 family proteins. An attractive therapeutic approach is to inhibit these proteins using agents that mimic the Bcl-2 homology 3 (BH3) domains of the proapoptotic Bcl-2 family members, which neutralize these proteins by binding to their surface hydrophobic grooves. A number of BH3 mimetic peptides and small molecules have been described, a few of which have advanced into clinical trials. Recent studies have highlighted ABT-737, a bona fide BH3 mimetic and potent inhibitor of antiapoptotic Bcl-2 family members, as a promising anticancer agent. This review summarizes recent advances in understanding the mechanisms of action of BH3 domains and several classes of BH3 mimetics, as well as the prospects of using these agents to improve cancer therapy.
Collapse
Affiliation(s)
- Lin Zhang
- University of Pittsburgh Cancer Institute, Departments of Pharmacology and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
134
|
Mitsiades CS, Hayden P, Kotoula V, McMillin DW, McMullan C, Negri J, Delmore JE, Poulaki V, Mitsiades N. Bcl-2 overexpression in thyroid carcinoma cells increases sensitivity to Bcl-2 homology 3 domain inhibition. J Clin Endocrinol Metab 2007; 92:4845-52. [PMID: 17848408 DOI: 10.1210/jc.2007-0942] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CONTEXT The Bcl-2 family of proteins regulates apoptosis in various models and may represent a promising therapeutic target in human malignancies. OBJECTIVE/METHODS We evaluated the sensitivity of thyroid carcinoma cell lines (two papillary, one follicular, two anaplastic, three medullary) in vitro to BH3I-1 and BH3I-2', two cell-permeable inhibitors of the Bcl-2 homology (BH)-3 domain-mediated interaction between proapoptotic and antiapoptotic Bcl-2 family members. The thyroid carcinoma cell line FRO was stably transfected with cDNA for Bcl-2 or constitutively active Akt and evaluated for sensitivity to BH3-domain inhibition. RESULTS BH3-domain inhibition disrupted the mitochondrial membrane potential in thyroid carcinoma cells, induced caspase-dependent apoptosis, and potently sensitized them to sublethal concentrations of doxorubicin and the proteasome inhibitor bortezomib (Velcade). Overexpression of constitutively active Akt suppressed BH3I-1-induced cell death. Bcl-2-overexpressing FRO cells were more resistant to conventional chemotherapeutic agents (such as doxorubicin) but significantly more sensitive to BH3I-1 than control cells and were found to overexpress caspase-9, caspase-8, Bmf, Bok, and Bik transcripts and express less A1, BRaf, and FLIP transcripts. CONCLUSIONS Bcl-2 expression protects thyroid carcinomas against chemotherapy-induced apoptosis. Nevertheless, overexpression of Bcl-2 may result in "oncogene addiction" of the cancer cell, which can be exploited by using BH3-domain inhibitors alone or in combination with other agents, including conventional chemotherapeutics (such as doxorubicin) or novel targeted therapies (such as the proteasome inhibitor bortezomib), for the treatment of aggressive thyroid cancer, including the medullary and anaplastic types.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana Farber Cancer Institute, Mayer Building, Room M555, 44 Binney Street, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Shoemaker AR, Oleksijew A, Bauch J, Belli BA, Borre T, Bruncko M, Deckwirth T, Frost DJ, Jarvis K, Joseph MK, Marsh K, McClellan W, Nellans H, Ng S, Nimmer P, O'Connor JM, Oltersdorf T, Qing W, Shen W, Stavropoulos J, Tahir SK, Wang B, Warner R, Zhang H, Fesik SW, Rosenberg SH, Elmore SW. A small-molecule inhibitor of Bcl-XL potentiates the activity of cytotoxic drugs in vitro and in vivo. Cancer Res 2007; 66:8731-9. [PMID: 16951189 DOI: 10.1158/0008-5472.can-06-0367] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inhibition of the prosurvival members of the Bcl-2 family of proteins represents an attractive strategy for the treatment of cancer. We have previously reported the activity of ABT-737, a potent inhibitor of Bcl-2, Bcl-X(L), and Bcl-w, which exhibits monotherapy efficacy in xenograft models of small-cell lung cancer and lymphoma and potentiates the activity of numerous cytotoxic agents. Here we describe the biological activity of A-385358, a small molecule with relative selectivity for binding to Bcl-X(L) versus Bcl-2 (K(i)'s of 0.80 and 67 nmol/L for Bcl-X(L) and Bcl-2, respectively). This compound efficiently enters cells and co-localizes with the mitochondrial membrane. Although A-385358 shows relatively modest single-agent cytotoxic activity against most tumor cell lines, it has an EC(50) of <500 nmol/L in cells dependent on Bcl-X(L) for survival. In addition, A-385358 enhances the in vitro cytotoxic activity of numerous chemotherapeutic agents (paclitaxel, etoposide, cisplatin, and doxorubicin) in several tumor cell lines. In A549 non-small-cell lung cancer cells, A-385358 potentiates the activity of paclitaxel by as much as 25-fold. Importantly, A-385358 also potentiated the activity of paclitaxel in vivo. Significant inhibition of tumor growth was observed when A-385358 was added to maximally tolerated or half maximally tolerated doses of paclitaxel in the A549 xenograft model. In tumors, the combination therapy also resulted in a significant increase in mitotic arrest followed by apoptosis relative to paclitaxel monotherapy.
Collapse
Affiliation(s)
- Alex R Shoemaker
- Global Pharmaceutical Research and Development, Abbott Laboratories, Abbott Park, Illinois 60064, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Zimmermann AK, Loucks FA, Schroeder EK, Bouchard RJ, Tyler KL, Linseman DA. Glutathione binding to the Bcl-2 homology-3 domain groove: a molecular basis for Bcl-2 antioxidant function at mitochondria. J Biol Chem 2007; 282:29296-304. [PMID: 17690097 PMCID: PMC2386251 DOI: 10.1074/jbc.m702853200] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bcl-2 protects cells against mitochondrial oxidative stress and subsequent apoptosis. However, the mechanism underlying the antioxidant function of Bcl-2 is currently unknown. Recently, Bax and several Bcl-2 homology-3 domain (BH3)-only proteins (Bid, Puma, and Noxa) have been shown to induce a pro-oxidant state at mitochondria (1-4). Given the opposing effects of Bcl-2 and Bax/BH3-only proteins on the redox state of mitochondria, we hypothesized that the antioxidant function of Bcl-2 is antagonized by its interaction with the BH3 domains of pro-apoptotic family members. Here, we show that BH3 mimetics that bind to a hydrophobic surface (the BH3 groove) of Bcl-2 induce GSH-sensitive mitochondrial dysfunction and apoptosis in cerebellar granule neurons. BH3 mimetics displace a discrete mitochondrial GSH pool in neurons and suppress GSH transport into isolated rat brain mitochondria. Moreover, BH3 mimetics and the BH3-only protein, Bim, inhibit a novel interaction between Bcl-2 and GSH in vitro. These results suggest that Bcl-2 regulates an essential pool of mitochondrial GSH and that this regulation may depend upon Bcl-2 directly interacting with GSH via the BH3 groove. We conclude that this novel GSH binding property of Bcl-2 likely plays a central role in its antioxidant function at mitochondria.
Collapse
Affiliation(s)
- Angela K Zimmermann
- Eleanor Roosevelt Institute, Department of Biological Sciences, University of Denver, Denver, Colorado 80208, USA
| | | | | | | | | | | |
Collapse
|
137
|
Zheng CH, Zhou YJ, Zhu J, Ji HT, Chen J, Li YW, Sheng CQ, Lu JG, Jiang JH, Tang H, Song YL. Construction of a three-dimensional pharmacophore for Bcl-2 inhibitors by flexible docking and the multiple copy simultaneous search method. Bioorg Med Chem 2007; 15:6407-17. [PMID: 17629704 DOI: 10.1016/j.bmc.2007.06.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 06/23/2007] [Accepted: 06/27/2007] [Indexed: 01/30/2023]
Abstract
B-Cell lymphoma-2 (Bcl-2) protein is a new promising target for anticancer drugs. A number of anticancer Bcl-2 inhibitors with diverse chemical structures have been discovered in recent years. In this paper, the flexible docking was performed to determine the binding modes of the representative inhibitors from different structural types. Subsequently, the binding modes of inhibitor were used to construct a primary three- dimensional (3D) pharmacophore model. It proved that this model can effectively disrupt the binding of the BH3 domain of proapoptotic Bcl-2 family members to Bcl-2, and match the structural requirement of a new type of Bcl-2 inhibitors. However, these distances between pharmacophoric points are not optimal due to the fact that not all of individual functional groups are located in the ideal position when inhibitors bind to its receptor. In this paper, we proposed a new idea to improve the quality of the pharmacophore model: the multiple copy simultaneous search (MCSS) method was performed to determine the energetically favorable distribution of functional groups with similar features to these pharmacophoric points in the active site of Bcl-2 first. Then their most energetically favorable minima in the positions near the pharmacophoric points were used to optimize the distances between pharmacophoric points. By examining the binding modes of several inhibitors from the same structural type, it was found that the more potent the inhibitor was, the closer it was to the optimized distances between pharmacophoric points. The optimized 3D pharmacophore model obtained in this paper may provide a good starting point for further rational design of Bcl-2 inhibitors.
Collapse
Affiliation(s)
- Can-Hui Zheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Jin L, Zhang Z, Wang Y, Wei M, Xu Q. A novel small molecule inhibitor targeted at Bcl-2. SCIENCE IN CHINA. SERIES C, LIFE SCIENCES 2007; 50:624-9. [PMID: 17879060 DOI: 10.1007/s11427-007-0079-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 06/05/2007] [Indexed: 10/22/2022]
Abstract
Recently, the heterocyclic compound 8-oxo-3-thiomorpholino-8H-acenaphtho[1,2-b]pyrrole-9-carbonitrile (S1) was synthesized and shown to induce apoptosis in both (H22) hematoma and (MCF-7) adenocarcinoma cells. The IC(50) values of S1 against the two cell lines were 0.17 and 0.09 micromol/L, respectively. Furthermore, the apoptosis-inducing activity of this compound was highlighted both in vivo and in vitro. Subsequent experiments identified Bcl-2 as the primary target of S1, as a significant reduction in Bcl-2 protein levels was observed in H22 cells following a two-hour treatment with 10 micromol/L S1. While rapid depolarization of mitochondrial membranes led immediately to caspase 9 activation, no changes were identified in either caspase 8 levels or levels in Bcl-2 mRNA. These data were consistent with the results of circular dichroism (CD) spectra analysis, revealing that S1 inactivated the Bcl-2 protein by destroying its critical alpha helices. Taken together, these results suggest the potential of S1 in the development of new therapeutic agents.
Collapse
Affiliation(s)
- LiJi Jin
- School of Environmental & Biological Science & Technology, Dalian University of Technology, Dalian 116024, China
| | | | | | | | | |
Collapse
|
139
|
Doshi JM, Tian D, Xing C. Ethyl-2-amino-6-bromo-4-(1-cyano-2-ethoxy-2-oxoethyl)-4H- chromene-3-carboxylate (HA 14-1), a Prototype Small-Molecule Antagonist against Antiapoptotic Bcl-2 Proteins, Decomposes To Generate Reactive Oxygen Species That Induce Apoptosis. Mol Pharm 2007; 4:919-28. [PMID: 17874842 DOI: 10.1021/mp7000846] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Overexpressing antiapoptotic Bcl-2 proteins to suppress apoptosis is one major mechanism via which cancer cells acquire drug resistance against cancer therapy. Ethyl-2-amino-6-bromo-4-(1-cyano-2-ethoxy-2-oxoethyl)-4 H-chromene-3-carboxylate (HA 14-1) is one of the earliest small-molecule antagonists against antiapoptotic Bcl-2 proteins. Since its discovery, HA 14-1 has been shown to be able to synergize a variety of anticancer agents. HA 14-1 also could selectively eliminate tumor cells with elevated level of Bcl-2 protein. HA 14-1, therefore, is being intensely investigated as a potential anticancer agent. Previous reports of HA 14-1 implied that it may not be stable, raising the question of whether HA 14-1 is a suitable drug candidate. The potential stability also raised the concern about whether HA 14-1 is the bioactive species. In this report, we confirm that HA 14-1 is not stable under physiological conditions: it rapidly decomposes in RPMI cell culture medium with a half-life of 15 min. This decomposition process also generates reactive oxygen species (ROS). To identify the actual candidate(s) for the observed bioactivity of HA 14-1, we characterized the structures, quantified the amount, and evaluated the bioactivities of the decomposed products. We also used ROS scavengers to explore the function of ROS. From these studies, we established that none of the decomposition products could account for the bioactivity of HA 14-1. ROS generated during the decomposition process, however, are critical for the in vitro cytotoxicity and the apoptosis induced by HA 14-1. This study demonstrates that HA 14-1 is not stable under physiological conditions and that HA 14-1 can generate ROS through its decomposition, independent of Bcl-2 antagonism. Because of its intrinsic tendency to decompose and to generate ROS, caution should be taken in using HA 14-1 as a qualified antagonist against antiapoptotic Bcl-2 proteins.
Collapse
Affiliation(s)
- Jignesh M Doshi
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 8-101 WDH, 308 Harvard Street SE, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
140
|
Sugaya N, Ikeda K, Tashiro T, Takeda S, Otomo J, Ishida Y, Shiratori A, Toyoda A, Noguchi H, Takeda T, Kuhara S, Sakaki Y, Iwayanagi T. An integrative in silico approach for discovering candidates for drug-targetable protein-protein interactions in interactome data. BMC Pharmacol 2007; 7:10. [PMID: 17705877 PMCID: PMC2045083 DOI: 10.1186/1471-2210-7-10] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Accepted: 08/20/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Protein-protein interactions (PPIs) are challenging but attractive targets for small chemical drugs. Whole PPIs, called the 'interactome', have been emerged in several organisms, including human, based on the recent development of high-throughput screening (HTS) technologies. Individual PPIs have been targeted by small drug-like chemicals (SDCs), however, interactome data have not been fully utilized for exploring drug targets due to the lack of comprehensive methodology for utilizing these data. Here we propose an integrative in silico approach for discovering candidates for drug-targetable PPIs in interactome data. RESULTS Our novel in silico screening system comprises three independent assessment procedures: i) detection of protein domains responsible for PPIs, ii) finding SDC-binding pockets on protein surfaces, and iii) evaluating similarities in the assignment of Gene Ontology (GO) terms between specific partner proteins. We discovered six candidates for drug-targetable PPIs by applying our in silico approach to original human PPI data composed of 770 binary interactions produced by our HTS yeast two-hybrid (HTS-Y2H) assays. Among them, we further examined two candidates, RXRA/NRIP1 and CDK2/CDKN1A, with respect to their biological roles, PPI network around each candidate, and tertiary structures of the interacting domains. CONCLUSION An integrative in silico approach for discovering candidates for drug-targetable PPIs was applied to original human PPIs data. The system excludes false positive interactions and selects reliable PPIs as drug targets. Its effectiveness was demonstrated by the discovery of the six promising candidate target PPIs. Inhibition or stabilization of the two interactions may have potential therapeutic effects against human diseases.
Collapse
Affiliation(s)
- Nobuyoshi Sugaya
- PharmaDesign, Inc., 2-19-8 Hatchobori, Chuo-ku, Tokyo, 104-0032, Japan
| | - Kazuyoshi Ikeda
- PharmaDesign, Inc., 2-19-8 Hatchobori, Chuo-ku, Tokyo, 104-0032, Japan
| | - Toshiyuki Tashiro
- PharmaDesign, Inc., 2-19-8 Hatchobori, Chuo-ku, Tokyo, 104-0032, Japan
| | - Shizu Takeda
- Central Research Laboratory, Hitachi, Ltd., 1-280 Higashi-koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Jun Otomo
- Central Research Laboratory, Hitachi, Ltd., 1-280 Higashi-koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Yoshiko Ishida
- Central Research Laboratory, Hitachi, Ltd., 1-280 Higashi-koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Akiko Shiratori
- Central Research Laboratory, Hitachi, Ltd., 1-280 Higashi-koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Atsushi Toyoda
- Genomic Sciences Center, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Hideki Noguchi
- Genomic Sciences Center, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Tadayuki Takeda
- Genomic Sciences Center, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Satoru Kuhara
- Graduate School of Genetic Resources Technology, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | - Yoshiyuki Sakaki
- Genomic Sciences Center, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takao Iwayanagi
- Research & Development Group, Hitachi, Ltd., 1-6-1 Marunouchi, Chiyoda-ku, Tokyo, 100-8220, Japan
| |
Collapse
|
141
|
Rega MF, Leone M, Jung D, Cotton NJ, Stebbins JL, Pellecchia M. Structure-based discovery of a new class of Bcl-xL antagonists. Bioorg Chem 2007; 35:344-53. [PMID: 17512966 PMCID: PMC2023964 DOI: 10.1016/j.bioorg.2007.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 03/02/2007] [Accepted: 03/03/2007] [Indexed: 11/22/2022]
Abstract
Apoptosis, or programmed cell death, plays a key role in normal tissue homeostasis ensuring a proper balance between cell production and cell loss. Anti-apoptotic Bcl-2-family proteins are central regulators of the apoptotic pathway and due to their ability to confer tumor resistance to chemotherapy or radiation, have been recently validated as targets for cancer drug discovery. Since the crucial interaction between pro- and anti-apoptotic members occurs via a conserved region located on the surface of the protein, a viable way to inhibit the anti-death activity of Bcl-2 proteins is to design small molecule inhibitors that occupy this cavity. Here, we describe a structure-based approach that led to the identification of four small molecule inhibitors directed at the hydrophobic groove on the surface of the Bcl-2 family protein Bcl-xL. The compounds were characterized in a number of assays including in vitro binding using 15N-labeled protein, a displacement DELFIA assay, and a cell-based viability assay with human cancer cells.
Collapse
Affiliation(s)
- Michele F. Rega
- Burnham Institute for Medical Research, Cancer Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
- Burnham Institute for Medical Research, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
- Department of Pharmaceutical Sciences, University of Salerno, ITALY
| | - Marilisa Leone
- Burnham Institute for Medical Research, Cancer Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Dawoon Jung
- Burnham Institute for Medical Research, Cancer Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Naomi J.H. Cotton
- Burnham Institute for Medical Research, Cancer Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
- Burnham Institute for Medical Research, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - John L. Stebbins
- Burnham Institute for Medical Research, Cancer Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Maurizio Pellecchia
- Burnham Institute for Medical Research, Cancer Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
- Burnham Institute for Medical Research, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| |
Collapse
|
142
|
Yang Z, Gagarin D, Ramezani A, Hawley RG, McCaffrey TA. Resistance to fas-induced apoptosis in cells from human atherosclerotic lesions: elevated Bcl-XL inhibits apoptosis and caspase activation. J Vasc Res 2007; 44:483-94. [PMID: 17657164 DOI: 10.1159/000106466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Accepted: 05/09/2007] [Indexed: 01/26/2023] Open
Abstract
The inappropriate survival of cells in the neointima contributes to atherosclerotic plaque progression, while apoptosis in the fibrous cap of lesions contributes to myocardial infarction and stroke. Prior genomic-scale transcript profiling of human carotid artery plaque cells with known sensitivity or resistance to fas-induced apoptosis identified candidate genes involved in lesion cell apoptosis. Retroviral overexpression indicated that several candidate factors were not causative, but that Bcl-X(L) conferred complete resistance to apoptosis induced by fas ligation. Resistant cells failed to efficiently activate caspase 8, an effect which was also observed in Bcl-X(L)-transfected cells. Small-molecule Bcl-2/X(L) inhibitors and siRNA knockdown of Bcl-X(L) markedly sensitized resistant cells to apoptosis, and partially restored caspase 8 activation. Caspase 3, 6 and 9 inhibitors reduced caspase 8 activation and blocked apoptosis. Complete knockdown of caspase 9 did not reduce apoptosis, while knockdown of Bid suppressed apoptosis, suggesting that mitochondrial pathways independent of caspase 9, such as Smac/Diablo or AIF, provide a necessary mitochondrial input to efficient caspase activation. Bcl-X(L) appears to modulate lesion cell apoptosis by suppressing mitochondrial amplification of caspase activation loops. The results may have direct implications for controlling plaque instability/progression, and identify a new class of small molecules to inhibit restenosis.
Collapse
Affiliation(s)
- Zhaoqing Yang
- Department of Biochemistry and Molecular Biology, George Washington University Medical Center, Washington, DC 20037, USA
| | | | | | | | | |
Collapse
|
143
|
Tang G, Ding K, Nikolovska-Coleska Z, Yang CY, Qiu S, Shangary S, Wang R, Guo J, Gao W, Meagher J, Stuckey J, Krajewski K, Jiang S, Roller PP, Wang S. Structure-based design of flavonoid compounds as a new class of small-molecule inhibitors of the anti-apoptotic Bcl-2 proteins. J Med Chem 2007; 50:3163-6. [PMID: 17552510 PMCID: PMC2527594 DOI: 10.1021/jm070383c] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Structure-based strategy was employed to design flavonoid compounds to mimic the Bim BH3 peptide as a new class of inhibitors of the anti-apoptotic Bcl-2 proteins. The most potent compound, 4 (BI-33), binds to Bcl-2 and Mcl-1 with Ki values of 17 and 18 nM, respectively. Compound 4 inhibits cell growth in the MDA-MB-231 breast cancer cell line with an IC50 value of 110 nM and effectively induces apoptosis.
Collapse
Affiliation(s)
- Guozhi Tang
- Comprehensive Cancer Center and Department of Internal Medicine, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Mohammad RM, Goustin AS, Aboukameel A, Chen B, Banerjee S, Wang G, Nikolovska-Coleska Z, Wang S, Al-Katib A. Preclinical studies of TW-37, a new nonpeptidic small-molecule inhibitor of Bcl-2, in diffuse large cell lymphoma xenograft model reveal drug action on both Bcl-2 and Mcl-1. Clin Cancer Res 2007; 13:2226-35. [PMID: 17404107 DOI: 10.1158/1078-0432.ccr-06-1574] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Overexpression of Bcl-2 protein has been observed in more than 80% of B-cell lymphomas, including diffuse large cell lymphoma (DLCL), the most common subtype of non-Hodgkin's lymphoma. We have previously employed the natural product (-)-gossypol to test its therapeutic potential as a small-molecule inhibitor of Bcl-2 for the treatment of B-cell lymphomas. EXPERIMENTAL DESIGN Recently, we have used a structure-based strategy to design a new class of potent small-molecule inhibitor acting on Bcl-2. One such lead compound is the benzenesulfonyl derivative TW-37, which was designed to target the BH3-binding groove in Bcl-2 where proapoptotic Bcl-2 proteins, such as Bak, Bax, Bid, and Bim bind. RESULTS In our fluorescence polarization-based binding assays using recombinant Bcl-2, Bcl-X(L), and Mcl-1 proteins, TW-37 binds to Bcl-2, Bcl-X(L), and Mcl-1 with K(i) values of 290, 1,110 and 260 nmol/L, respectively. Hence, TW-37 is a potent inhibitor of Bcl-2 and has >3-fold selectivity over Bcl-X(L). In vitro, TW-37 showed significant antiproliferative effect in a de novo chemoresistant WSU-DLCL(2) lymphoma cell line and primary cells obtained from a lymphoma patient with no effect on normal peripheral blood lymphocytes. Coimmunoprecipitation experiments showed that TW-37 disrupted heterodimer formation between Bax or truncated-Bid and antiapoptotic proteins in the order Mcl-1 > Bcl-2 >> Bcl-X(L). As expected, TW-37 caused apoptotic death. Pre-exposure of lymphoma cells to TW-37 significantly enhanced the killing effect of cyclophosphamide-doxorubicin-vincristine-prednisone (CHOP) regimen. The maximum tolerated dose of TW-37 in severe combined immunodeficient (SCID) mice was 40 mg/kg for three i.v. injections when given alone and 20 mg/kg, x3 when given in combination with CHOP. Using WSU-DLCL(2)-SCID mouse xenograft model, the addition of TW-37 to CHOP resulted in more complete tumor inhibition compared with either CHOP or TW-37 alone. CONCLUSIONS We conclude that the administration of TW-37, as a potent Bcl-2 and Mcl-1 inhibitor, to standard chemotherapy may prove an effective strategy in the treatment of B-cell lymphoma.
Collapse
Affiliation(s)
- Ramzi M Mohammad
- Division of Hematology and Oncology, Department of Internal Medicine, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Sadowsky JD, Murray JK, Tomita Y, Gellman SH. Exploration of Backbone Space in Foldamers Containing α- and β-Amino Acid Residues: Developing Protease-Resistant Oligomers that Bind Tightly to the BH3-Recognition Cleft of Bcl-xL. Chembiochem 2007; 8:903-16. [PMID: 17503422 DOI: 10.1002/cbic.200600546] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein-protein interactions play crucial roles in cell-signaling events and are often implicated in human disease. Molecules that bind tightly to functional protein-surface sites and show high stability to degradative enzymes could be valuable pharmacological tools for dissection of cell-signaling networks and might ultimately lead to therapeutic agents. We recently described oligomers containing both alpha- and beta-amino acid residues that bind tightly to the BH3 recognition site of the anti-apoptotic protein Bcl-x(L). The oligomers with highest affinity had a nine-residue N-terminal segment with a 1:1 alpha:beta residue repeat and a six-residue C-terminal segment containing exclusively proteinogenic alpha-residues. The N-terminal portions of such (alpha/beta+alpha)-peptides are highly resistant to proteolysis, but the C-terminal alpha-segments are susceptible. This study emerged from efforts to modify the alpha-segment in an (alpha/beta+alpha)-peptide in a way that would diminish proteolytic degradation but retain high affinity for Bcl-x(L). Some of the oligomers reported here could prove useful in certain biological applications, particularly those for which extended incubation in a biological milieu is required.
Collapse
Affiliation(s)
- Jack D Sadowsky
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
146
|
Tang G, Yang CY, Nikolovska-Coleska Z, Guo J, Qiu S, Wang R, Gao W, Wang G, Stuckey J, Krajewski K, Jiang S, Roller PP, Wang S. Pyrogallol-based molecules as potent inhibitors of the antiapoptotic Bcl-2 proteins. J Med Chem 2007; 50:1723-6. [PMID: 17378545 PMCID: PMC2536619 DOI: 10.1021/jm061400l] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report herein a new class of small-molecule inhibitors of antiapoptotic Bcl-2 proteins. The most potent compound, 7, binds to Bcl-2, Bcl-xL, and Mcl-1 proteins with Ki of 110, 638, and 150 nM, respectively. Compound 7 is highly effective in induction of cell death in breast cancer cells with high levels of Bcl-2, Bcl-xL, and Mcl-1 proteins and represents a promising lead compound for the design of new anticancer drugs.
Collapse
Affiliation(s)
- Guozhi Tang
- Department of Internal Medicine, Comprehensive Cancer Center, and Life Sciences Institute, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Zhang M, Ling Y, Yang CY, Liu H, Wang R, Wu X, Ding K, Zhu F, Griffith BN, Mohammad RM, Wang S, Yang D. A novel Bcl-2 small molecule inhibitor 4-(3-methoxy-phenylsulfannyl)-7-nitro-benzofurazan-3-oxide (MNB)-induced apoptosis in leukemia cells. Ann Hematol 2007; 86:471-81. [PMID: 17396262 DOI: 10.1007/s00277-007-0288-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 03/12/2007] [Indexed: 10/23/2022]
Abstract
A novel small molecule inhibitor, 4-(3-methoxy-phenylsulfannyl)-7-nitro-benzofurazan-3-oxide (MNB), competes with the Bak BH3 peptide to bind Bcl-2 protein with a binding affinity of IC(50) = 0.70 microM, as assessed by a fluorescence polarization based binding assay. HL-60 cells express the highest levels of Bcl-2 among the cell lines examined. Treated with 5 microM of MNB only for 6 h, 85% of HL-60 cells were detected to undergo apoptosis. Pan-caspase inhibitor, Z-VAD-FMK, blocks MNB-induced apoptosis in HL-60 cells. Caspase-2, caspase-3, caspase-7, caspase-8, caspase-9, and PARP activation were observed at as early as 4 to 6 h of MNB treatment. In addition, it has been confirmed that the caspase-3 specific inhibitor, Z-DEVD-FMK, blocks the activation of caspase-8 in MNB-treated HL-60 cells. MNB treatment does not change Bcl-2 or Bax expression level in HL-60 cells, but causes Bid cleavage. Further experiments have illustrated that MNB inhibits the heterodimerization of Bcl-2 with Bax or Bid, reduces the mitochondrial membrane potential (DeltaPsimt), and induces cytochrome c release from mitochondria in HL-60 cells. These results suggest that MNB induces apoptosis in HL-60 by inhibiting the heterodimerization of Bcl-2 with pro-apoptosis Bcl-2 members, resulting in a decrease in the mitochondrial membrane potential and cytochrome c release, activation of caspases and PARP; it is a caspase-dependent process in which the activation of caspase-8 is dependent on the mitochondrial apoptosis signal transduction pathway. MNB prolongs the life spans of HL-60 bearing mice, potently kills fresh AML and ALL cells, indicating that it has the potential to be developed to treat leukemia.
Collapse
Affiliation(s)
- Manchao Zhang
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Coluccia AML, Gunby RH, Tartari CJ, Scapozza L, Gambacorti-Passerini C, Passoni L. Anaplastic lymphoma kinase and its signalling molecules as novel targets in lymphoma therapy. Expert Opin Ther Targets 2007; 9:515-32. [PMID: 15948671 DOI: 10.1517/14728222.9.3.515] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A crucial issue in the development of molecularly-targeted anticancer therapies is the identification of appropriate molecules whose targeting would result in tumour regression with a minimal level of systemic toxicity. Anaplastic lymphoma kinase (ALK) is a transmembrane receptor tyrosine kinase, normally expressed at low levels in the nervous system. As a consequence of chromosomal translocations involving the alk gene (2p23), ALK is also aberrantly expressed and constitutively activated in approximately 60% of CD30+ anaplastic large cell lymphomas (ALCLs). Due to the selective overexpression of ALK in tumour cells, its direct involvement in the process of malignant transformation and its frequent expression in ALCL patients, the authors recognise ALK as a suitable candidate for the development of molecularly targeted strategies for the therapeutic treatment of ALK-positive lymphomas. Strategies targeting ALK directly or indirectly via the inhibition of the protein networks responsible for ALK oncogenic signalling are discussed.
Collapse
Affiliation(s)
- A M L Coluccia
- Oncogenic Fusion Genes and Proteins Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | | | | | | | | | | |
Collapse
|
149
|
Liu Z, Larock RC. Synthesis of Carbazoles and Dibenzofurans via Cross-Coupling of o-Iodoanilines and o-Iodophenols with Silylaryl Triflates and Subsequent Pd-Catalyzed Cyclization. Tetrahedron 2007; 63:347-355. [PMID: 18180808 PMCID: PMC2084200 DOI: 10.1016/j.tet.2006.10.071] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
An efficient route to a variety of carbazoles and dibenzofurans has been developed. It involves the reaction of o-iodoanilines or o-iodophenols with silylaryl triflates in the presence of CsF to afford the N- or O-arylated products, which are subsequently cyclized using a Pd catalyst to carbazoles and dibenzofurans in good to excellent yields. By using this methodology, the carbazole alkaloid mukonine has been synthesized in 76% overall yield in three steps.
Collapse
Affiliation(s)
- Zhijian Liu
- Department of Chemistry, Iowa State University, Ames, Iowa 50011
| | | |
Collapse
|
150
|
Abstract
Proteins in nature fold into native conformations in which combinations of peripherally projected aliphatic, aromatic and ionic functionalities direct a wide range of properties. Alpha-helices, one of the most common protein secondary structures, serve as important recognition regions on protein surfaces for numerous protein-protein, protein-DNA and protein-RNA interactions. These interactions are characterized by conserved structural features within the alpha-helical domain. Rational design of structural mimetics of these domains with synthetic small molecules has proven an effective means to modulate such protein functions. In this tutorial review we discuss strategies that utilize synthetic small-molecule antagonists to selectively target essential protein-protein interactions involved in certain diseases. We also evaluate some of the protein-protein interactions that have been or are potential targets for alpha-helix mimetics.
Collapse
Affiliation(s)
- Jessica M Davis
- Department of Chemistry, Fairfield University, Fairfield, CT 06824, USA.
| | | | | |
Collapse
|