151
|
Barbosa DJ, Capela JP, Feio-Azevedo R, Teixeira-Gomes A, Bastos MDL, Carvalho F. Mitochondria: key players in the neurotoxic effects of amphetamines. Arch Toxicol 2015; 89:1695-1725. [PMID: 25743372 DOI: 10.1007/s00204-015-1478-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/09/2015] [Indexed: 12/21/2022]
Abstract
Amphetamines are a class of psychotropic drugs with high abuse potential, as a result of their stimulant, euphoric, emphathogenic, entactogenic, and hallucinogenic properties. Although most amphetamines are synthetic drugs, of which methamphetamine, amphetamine, and 3,4-methylenedioxymethamphetamine ("ecstasy") represent well-recognized examples, the use of natural related compounds, namely cathinone and ephedrine, has been part of the history of humankind for thousands of years. Resulting from their amphiphilic nature, these drugs can easily cross the blood-brain barrier and elicit their well-known psychotropic effects. In the field of amphetamines' research, there is a general consensus that mitochondrial-dependent pathways can provide a major understanding concerning pathological processes underlying the neurotoxicity of these drugs. These events include alterations on tricarboxylic acid cycle's enzymes functioning, inhibition of mitochondrial electron transport chain's complexes, perturbations of mitochondrial clearance mechanisms, interference with mitochondrial dynamics, as well as oxidative modifications in mitochondrial macromolecules. Additionally, other studies indicate that amphetamines-induced neuronal toxicity is closely regulated by B cell lymphoma 2 superfamily of proteins with consequent activation of caspase-mediated downstream cell death pathway. Understanding the molecular mechanisms at mitochondrial level involved in amphetamines' neurotoxicity can help in defining target pathways or molecules mediating these effects, as well as in developing putative therapeutic approaches to prevent or treat the acute- or long-lasting neuropsychiatric complications seen in human abusers.
Collapse
Affiliation(s)
- Daniel José Barbosa
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.
| | - João Paulo Capela
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
- FP-ENAS (Unidade de Investigação UFP em energia, Ambiente e Saúde), CEBIMED (Centro de Estudos em Biomedicina), Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Rua 9 de Abril 349, 4249-004, Porto, Portugal
| | - Rita Feio-Azevedo
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Armanda Teixeira-Gomes
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
152
|
Abstract
Cell death and inflammation are ancient processes of fundamental biological importance in both normal physiology and human disease pathologies. The recent observation that apoptosis regulatory components have dual roles in cell death and inflammation suggests that these proteins function, not primarily to kill, but to coordinate tissue repair and remodeling. This perspective unifies cell death components as positive regulators of tissue repair that replaces malfunctioning or damaged tissues and enhances the resilience of epithelia to insult. It is now recognized that cells that die by apoptosis do not do so silently, but release a variety of paracrine signals to communicate with their cellular environment to ensure tissue regeneration, and wound healing. Moreover, inflammatory signaling pathways, such as those emanating from the TNF receptor or Toll-related receptors, take part in cell competition to eliminate developmentally aberrant clones. Ubiquitylation has emerged as crucial mediator of signal transduction in cell death and inflammation. Here, we focus on recent advances on ubiquitin-mediated regulation of cell death and inflammation, and how this is used to regulate the defense of homeostasis.
Collapse
|
153
|
Abstract
Inhibitors of apoptosis (IAPs) family of genes encode baculovirus IAP-repeat domain-containing proteins with antiapoptotic function. These proteins also contain RING or UBC domains and act by binding to major proapoptotic factors and ubiquitylating them. High levels of IAPs inhibit caspase-mediated apoptosis. For these cells to undergo apoptosis, IAP function must be neutralized by IAP-antagonists. Mammalian IAP knockouts do not exhibit obvious developmental phenotypes, but the cells are more sensitized to apoptosis in response to injury. Loss of the mammalian IAP-antagonist ARTS results in reduced stem cell apoptosis. In addition to the antiapoptotic properties, IAPs regulate the innate immune response, and the loss of IAP function in humans is associated with immunodeficiency. The roles of IAPs in Drosophila apoptosis regulation are more apparent, where the loss of IAP1, or the expression of IAP-antagonists in Drosophila cells, is sufficient to trigger apoptosis. In this organism, apoptosis as a fate is conferred by the transcriptional induction of the IAP-antagonists. Many signaling pathways often converge on shared enhancer regions of IAP-antagonists. Cell death sensitivity is further regulated by posttranscriptional mechanisms, including those regulated by kinases, miRs, and ubiquitin ligases. These mechanisms are employed to eliminate damaged or virus-infected cells, limit neuroblast (neural stem cell) numbers, generate neuronal diversity, and sculpt tissue morphogenesis.
Collapse
Affiliation(s)
- Deepika Vasudevan
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA.
| |
Collapse
|
154
|
Romero A, Novoa B, Figueras A. The complexity of apoptotic cell death in mollusks: An update. FISH & SHELLFISH IMMUNOLOGY 2015; 46:79-87. [PMID: 25862972 DOI: 10.1016/j.fsi.2015.03.038] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/28/2015] [Accepted: 03/07/2015] [Indexed: 06/04/2023]
Abstract
Apoptosis is a type of programmed cell death that produces changes in cell morphology and in biochemical intracellular processes without inflammatory reactions. The components of the apoptotic pathways are conserved throughout evolution. Caspases are key molecules involved in the transduction of the death signal and are responsible for many of the biochemical and morphological changes associated with apoptosis. Nowadays, It is known that caspases are activated through two major apoptotic pathways (the extrinsic or death receptor pathway and the intrinsic or mitochondrial pathway), but there are also evidences of at least other alternative pathway (the perforin/granzyme pathway). Apoptosis in mollusks seems to be similar in complexity to apoptosis in vertebrates but also has unique features maybe related to their recurrent exposure to environmental changes, pollutants, pathogens and also related to the sedentary nature of some stages in the life cycle of mollusks bivalves and gastropods. As in other animals, apoptotic process is involved in the maintenance of tissue homeostasis and also constitutes an important immune response that can be triggered by a variety of stimuli, including cytokines, hormones, toxic insults, viruses, and protozoan parasites. The main goal of this work is to present the current knowledge of the molecular mechanisms of apoptosis in mollusks and to highlight those steps that need further study.
Collapse
Affiliation(s)
- A Romero
- Instituto de Investigaciones Marinas, Consejo Superior de Investigaciones Científicas (CSIC), Eduardo Cabello 6, 36208 Vigo, Spain
| | - B Novoa
- Instituto de Investigaciones Marinas, Consejo Superior de Investigaciones Científicas (CSIC), Eduardo Cabello 6, 36208 Vigo, Spain
| | - A Figueras
- Instituto de Investigaciones Marinas, Consejo Superior de Investigaciones Científicas (CSIC), Eduardo Cabello 6, 36208 Vigo, Spain.
| |
Collapse
|
155
|
Talbott RL, Borzilleri RM, Chaudhry C, Fargnoli J, Shen H, Fairchild C, Barnhart B, Ortega M, McDonagh TE, Vuppugalla R, Vite GD, Hunt JT, Gottardis M, Naglich JG. Pharmacology of smac mimetics; chemotype differentiation based on physical association with caspase regulators and cellular transport. Exp Cell Res 2015; 338:251-60. [PMID: 26302264 DOI: 10.1016/j.yexcr.2015.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 01/08/2023]
Abstract
Cellular levels of inhibitor of apoptosis (IAP) proteins are elevated in multiple human cancers and their activities often play a part in promoting cancer cell survival by blocking apoptotic pathways, controlling signal transduction pathways and contributing to resistance. These proteins function through interactions of their BIR (baculoviral IAP repeat) protein domains with pathway components and these interactions are endogenously antagonized by Smac/Diablo (second mitochondrial activator of caspases/direct IAP binding protein with low isoelectric point). This report describes development of synthetic smac mimetics (SM) and compares their binding, antiproliferative and anti-tumor activities. All dimeric antagonists inhibit in vitro smac tetrapeptide binding to recombinant IAP proteins, rescue IAP-bound caspase-3 activity and show anti-proliferative activity against human A875 melanoma cells. One heterodimeric SM, SM3, binds tightly to IAP proteins in vitro and slowly dissociates (greater than two hours) from these protein complexes compared to the other antagonists. In addition, in vitro SM anti-proliferation potency is influenced by ABCB1 transporter (ATP-binding cassette, sub-family B; MDR1, P-gp) activities and one antagonist, SM5, does not appear to be an ABCB1 efflux pump substrate. All dimeric smac mimetics inhibit the growth of human melanoma A875 tumors implanted in athymic mice at well-tolerated doses. One antagonist, SM4, shows broad spectrum in vivo anti-tumor activity and modulates known pharmacodynamic markers of IAP antagonism. These data taken together demonstrate the range of diverse dimeric IAP antagonist activities and supports their potential as anticancer agents.
Collapse
Affiliation(s)
- Randy L Talbott
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA.
| | | | - Charu Chaudhry
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Joseph Fargnoli
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Henry Shen
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Craig Fairchild
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Bryan Barnhart
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Marie Ortega
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Thomas E McDonagh
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Ragini Vuppugalla
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Gregory D Vite
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - John T Hunt
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Marco Gottardis
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Joseph G Naglich
- Bristol-Myers Squibb Research, PO Box 4000, Princeton, NJ 08543-4000, USA
| |
Collapse
|
156
|
Chessari G, Buck IM, Day JEH, Day PJ, Iqbal A, Johnson CN, Lewis EJ, Martins V, Miller D, Reader M, Rees DC, Rich SJ, Tamanini E, Vitorino M, Ward GA, Williams PA, Williams G, Wilsher NE, Woolford AJA. Fragment-Based Drug Discovery Targeting Inhibitor of Apoptosis Proteins: Discovery of a Non-Alanine Lead Series with Dual Activity Against cIAP1 and XIAP. J Med Chem 2015. [PMID: 26218264 DOI: 10.1021/acs.jmedchem.5b00706] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inhibitor of apoptosis proteins (IAPs) are important regulators of apoptosis and pro-survival signaling pathways whose deregulation is often associated with tumor genesis and tumor growth. IAPs have been proposed as targets for anticancer therapy, and a number of peptidomimetic IAP antagonists have entered clinical trials. Using our fragment-based screening approach, we identified nonpeptidic fragments binding with millimolar affinities to both cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP). Structure-based hit optimization together with an analysis of protein-ligand electrostatic potential complementarity allowed us to significantly increase binding affinity of the starting hits. Subsequent optimization gave a potent nonalanine IAP antagonist structurally distinct from all IAP antagonists previously reported. The lead compound had activity in cell-based assays and in a mouse xenograft efficacy model and represents a highly promising start point for further optimization.
Collapse
Affiliation(s)
- Gianni Chessari
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Ildiko M Buck
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - James E H Day
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Philip J Day
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Aman Iqbal
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Christopher N Johnson
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Edward J Lewis
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Vanessa Martins
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Darcey Miller
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Michael Reader
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - David C Rees
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Sharna J Rich
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Emiliano Tamanini
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Marc Vitorino
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - George A Ward
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Pamela A Williams
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Glyn Williams
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Nicola E Wilsher
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Alison J-A Woolford
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| |
Collapse
|
157
|
Ikeda M, Kawakami M, Kitade Y. Pro-apoptotic Peptide Amphiphile Self-assembled with the Assistance of Polycations. CHEM LETT 2015. [DOI: 10.1246/cl.150399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Masato Ikeda
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University
- Department of Biomolecular Science, Graduate School of Engineering, Gifu University
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University
| | - Maika Kawakami
- Department of Biomolecular Science, Graduate School of Engineering, Gifu University
| | - Yukio Kitade
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University
- Department of Biomolecular Science, Graduate School of Engineering, Gifu University
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University
| |
Collapse
|
158
|
Abstract
Tumour necrosis factor (TNF) is a pro-inflammatory cytokine that has important roles in mammalian immunity and cellular homeostasis. Deregulation of TNF receptor (TNFR) signalling is associated with many inflammatory disorders, including various types of arthritis and inflammatory bowel disease, and targeting TNF has been an effective therapeutic strategy in these diseases. This Review focuses on the recent advances that have been made in understanding TNFR signalling and the consequences of its deregulation for cellular survival, apoptosis and regulated necrosis. We discuss how TNF-induced survival signals are distinguished from those that lead to cell death. Finally, we provide a brief overview of the role of TNF in inflammatory and autoimmune diseases, and we discuss up-to-date and future treatment strategies for these disorders.
Collapse
|
159
|
Chaudhary AK, Yadav N, Bhat TA, O'Malley J, Kumar S, Chandra D. A potential role of X-linked inhibitor of apoptosis protein in mitochondrial membrane permeabilization and its implication in cancer therapy. Drug Discov Today 2015; 21:38-47. [PMID: 26232549 DOI: 10.1016/j.drudis.2015.07.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/09/2015] [Accepted: 07/21/2015] [Indexed: 12/17/2022]
Abstract
X-chromosome-linked inhibitor of apoptosis protein (XIAP) has an important regulatory role in programmed cell death by inhibiting the caspase cascade. Activation of XIAP-dependent signaling culminates into regulation of multiple cellular processes including apoptosis, innate immunity, epithelial-to-mesenchymal transition, cell migration, invasion, metastasis and differentiation. Although XIAP localizes to the cytosolic compartment, XIAP-mediated cellular signaling encompasses mitochondrial and post-mitochondrial levels. Recent findings demonstrate that XIAP also localizes to mitochondria and regulates mitochondria functions. XIAP acts upstream of mitochondrial cytochrome c release and modulates caspase-dependent apoptosis. The new function of XIAP has potential to enhance mitochondrial membrane permeabilization and other cellular functions controlling cytochrome c release. These findings could exploit the overexpression of XIAP in human tumors for therapeutic benefits.
Collapse
Affiliation(s)
- Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Tariq A Bhat
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|
160
|
Elsayed NM, Abou El Ella DA, Serya RA, Abouzid KA. Targeting apoptotic machinery as approach for anticancer therapy: Smac mimetics as anticancer agents. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2015. [DOI: 10.1016/j.fjps.2015.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
161
|
Hird AW, Aquila BM, Hennessy EJ, Vasbinder MM, Yang B. Small molecule inhibitor of apoptosis proteins antagonists: a patent review. Expert Opin Ther Pat 2015; 25:755-74. [PMID: 25980951 DOI: 10.1517/13543776.2015.1041922] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The family of inhibitor of apoptosis proteins (IAPs) plays a key role in the suppression of proapoptotic signaling; hence, a small molecule that disrupts the binding of IAPs with their functional partner should restore apoptotic response to proapoptotic stimuli in cells. The continued publication of new patent applications of IAP antagonists over the past 4 years is a testament to the continued interest surrounding the IAP family of proteins. AREAS COVERED This review summarizes the IAP antagonist patent literature from 2010 to 2014. Monovalent and bivalent Smac mimetics will be covered as well as two new developments in the field: IAP antagonists coupled to or merged with other targeted agents and new BIR2 selective IAP antagonists. EXPERT OPINION In addition to the well-explored scaffolds for monovalent and bivalent Smac-mimetics, some companies have taken more drastic approaches to explore new chemical space - for example, fragment-based approaches and macrocyclic inhibitors. Furthermore, other companies have designed compounds with alternative biological profiles - tethering to known kinase binding structures, trying to target to the mitochondria or introducing selective binding to the BIR2 domain. An overview of the status for the four small molecule IAP antagonists being evaluated in active human clinical trials is also provided.
Collapse
Affiliation(s)
- Alexander W Hird
- AstraZeneca, Medicinal Chemistry, Oncology iMed , 35 Gatehouse Drive, Waltham, MA 02451 , USA +1 781 839 4145 ;
| | | | | | | | | |
Collapse
|
162
|
Qian Z, Xu X, Amacher JF, Madden DR, Cormet-Boyaka E, Pei D. Intracellular Delivery of Peptidyl Ligands by Reversible Cyclization: Discovery of a PDZ Domain Inhibitor that Rescues CFTR Activity. Angew Chem Int Ed Engl 2015; 54:5874-8. [PMID: 25785567 PMCID: PMC4424104 DOI: 10.1002/anie.201411594] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/23/2015] [Indexed: 12/13/2022]
Abstract
A general strategy was developed for the intracellular delivery of linear peptidyl ligands through fusion to a cell-penetrating peptide and cyclization of the fusion peptides via a disulfide bond. The resulting cyclic peptides are cell permeable and have improved proteolytic stability. Once inside the cell, the disulfide bond is reduced to produce linear biologically active peptides. This strategy was applied to generate a cell-permeable peptide substrate for real-time detection of intracellular caspase activities during apoptosis and an inhibitor for the CFTR-associated ligand (CAL) PDZ domain as a potential treatment for cystic fibrosis.
Collapse
Affiliation(s)
- Ziqing Qian
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, OH 43210 (USA)
| | - Xiaohua Xu
- Department of Veterinary Biosciences, The Ohio State University, 1950 Coffey Road, Columbus, OH 43210, (USA)
| | - Jeanine F. Amacher
- Department of Biochemistry, Dartmouth Medical School, 7200 Vail Building, Hanover, NH 03755 (USA)
| | - Dean R. Madden
- Department of Biochemistry, Dartmouth Medical School, 7200 Vail Building, Hanover, NH 03755 (USA)
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, 1950 Coffey Road, Columbus, OH 43210, (USA)
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, OH 43210 (USA)
| |
Collapse
|
163
|
Khashper A, Lubell WD. Design, synthesis, conformational analysis and application of indolizidin-2-one dipeptide mimics. Org Biomol Chem 2015; 12:5052-70. [PMID: 24899358 DOI: 10.1039/c4ob00777h] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Growth in the field of peptide mimicry over the past few decades has resulted in the synthesis of many new compounds and the investigation of novel pharmacological agents. Azabicyclo[X.Y.0]alkanone amino acids are among the attractive classes of constrained mimics, because they can create rigid peptide structures for probing the conformation and roles of natural motifs in recognition events important for biological activity. Herein, we review the last ten years of the synthesis, conformational analysis and activity of analogs of the azabicyclo[4.3.0]alkan-2-one amino acid subclass, so-called indolizidin-2-one amino acids, with particular attention on their employment as inputs for biological applications.
Collapse
Affiliation(s)
- Arkady Khashper
- Département de Chimie, Université de Montréal, Montréal H3C 3J7, Canada.
| | | |
Collapse
|
164
|
Li M, Liu P, Gao G, Deng J, Pan Z, Wu X, Xie G, Yue C, Cho CH, Ma Y, Cai L. Smac therapeutic Peptide nanoparticles inducing apoptosis of cancer cells for combination chemotherapy with Doxorubicin. ACS APPLIED MATERIALS & INTERFACES 2015; 7:8005-8012. [PMID: 25815797 DOI: 10.1021/acsami.5b00329] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Smac-conjugated nanoparticle (Smac-NP) was designed to induce the apoptosis of cancer cells and as a drug carrier for combination therapy. It contained three parts, a SmacN7 peptide which could induce apoptosis of cancer cells by interacting with XIAPs, the cell penetrating domain rich in arginine, and four hydrophobic tails for self-assembled Smac-NP. We demonstrated that Smac-NPs exerted an antitumor effect in breast cancer cell MDA-MB-231 and nonsmall lung cancer (NSCLC) cell H460, which efficiently inhibited cancer cells proliferation without influencing normal liver cell lines LO2. Smac-NPs also significantly induced apoptosis of MDA-MB-231 and H460 cells through activating pro-caspase-3, down-regulating the expression of antiapoptotic protein Bcl-2 and up-regulating the pro-apoptotic protein Bax. Furthermore, Smac-NPs could be explored as a drug delivery system to load hydrophobic drug such as DOX for combination therapy. The DOX-loaded nanoparticles (DOX-Smac-NPs) exhibited higher cellular uptake efficiency and antitumor effect. Our work provided a new insight into therapeutic peptides integrated with drug simultaneously in one system for cancer combination treatment.
Collapse
Affiliation(s)
- Mingxing Li
- †School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Peng Liu
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Guanhui Gao
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jizhe Deng
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhengyin Pan
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xu Wu
- †School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Gaofeng Xie
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Caixia Yue
- §Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chi Hin Cho
- †School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Yifan Ma
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lintao Cai
- ‡Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
165
|
Wazir U, Orakzai MMAW, Khanzada ZS, Jiang WG, Sharma AK, Kasem A, Mokbel K. The role of death-associated protein 3 in apoptosis, anoikis and human cancer. Cancer Cell Int 2015; 15:39. [PMID: 25883535 PMCID: PMC4399419 DOI: 10.1186/s12935-015-0187-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 03/18/2015] [Indexed: 01/12/2023] Open
Abstract
Death-associated protein 3 (DAP3) is a molecule with a significant role in the control of both apoptosis and anoikis. Apoptosis is the predominant type of programmed cell death (PCD) which may occur in response to irreparable damage to DNA, or in response to induction by inflammatory cells. Anoikis is subset of apoptosis which occurs in epithelial cells in response to detachment from the surrounding matrix. Both apoptosis and anoikis are of interest in the context of carcinogenesis. In this review, we shall discuss apoptosis and anoikis, and the recent literature regarding the role of DAP3 in both these pathways.
Collapse
Affiliation(s)
- Umar Wazir
- />The London Breast Institute, Princess Grace Hospital, London, UK
- />Department of Breast Surgery, St. George’s Hospital and Medical School, University of London, London, UK
| | | | - Zubair S Khanzada
- />Metastasis and Angiogenesis Research Group, University Department of Surgery, Cardiff University School of Medicine, Cardiff University, Cardiff, Wales UK
| | - Wen G Jiang
- />Metastasis and Angiogenesis Research Group, University Department of Surgery, Cardiff University School of Medicine, Cardiff University, Cardiff, Wales UK
| | - Anup K Sharma
- />Department of Breast Surgery, St. George’s Hospital and Medical School, University of London, London, UK
| | - Abdul Kasem
- />The London Breast Institute, Princess Grace Hospital, London, UK
| | - Kefah Mokbel
- />The London Breast Institute, Princess Grace Hospital, London, UK
- />Department of Breast Surgery, St. George’s Hospital and Medical School, University of London, London, UK
| |
Collapse
|
166
|
Qian Z, Xu X, Amacher JF, Madden DR, Cormet-Boyaka E, Pei D. Intracellular Delivery of Peptidyl Ligands by Reversible Cyclization: Discovery of a PDZ Domain Inhibitor that Rescues CFTR Activity. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201411594] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
167
|
Tyrosine kinase inhibitor tyrphostin AG490 retards chronic joint inflammation in mice. Inflammation 2015; 37:995-1005. [PMID: 24473905 DOI: 10.1007/s10753-014-9820-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tyrphostin AG490 is a Janus kinase (JAK) 2 inhibitor that is clinically used as an anticancer agent and is also effective in various models of inflammatory and autoimmune diseases. In this study, we examined the effects of tyrphostin AG490 on the development of collagenase-induced osteoarthritis (CIOA). Our results showed that tyrphostin-ameliorated cartilage and bone destructions. This effect was associated with decreased expression of signal transducers and activators of transcription 3 (STAT3), phosphorylated JAK2, Dickkopf homolog 1, and receptor activator of nuclear factor κB ligand (RANKL) in the joints of arthritic mice. Tyrphostin AG490 suppressed STAT3 phosphorylation and the expression of tumor necrosis factor-related apoptosis-inducing ligand and RANKL by synovial fluid cells. The drug inhibited RANKL-induced osteoclast differentiation in vitro. Molecules, such as tyrphostin AG490 that limit bone erosion and influence osteoclast generation, might have therapeutic utility in joint degenerative disorders.
Collapse
|
168
|
Xu Z, Chen Y, Xu G, Peng C, Liu E, Li Y, Niu J, Li C. Omi/HtrA2 pro-apoptotic marker differs in various hepatocellular carcinoma cell lines owing to ped/pea-15 expression level. Oncol Rep 2015; 33:905-12. [PMID: 25484138 DOI: 10.3892/or.2014.3656] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/08/2014] [Indexed: 11/05/2022] Open
Abstract
Omi/HtrA2 promotes cell apoptosis in human cancer cells. Early studies showed that primary hepatocellular carcinoma requires Omi/HtrA2 expression for cell apoptosis. Additionally, the Omi/HtrA2 pro-apoptotic marker demonstrated a difference in some cell types. However, how the Omi/HtrA2 pro-apoptotic marker reacts during the process of hepatocellular carcinoma cell apoptosis remains to be determined. Thus, we investigated the role and possible mechanism of Omi/HtrA2 on hepatocellular carcinoma cell apoptosis using various hepatocellular carcinoma cell lines. The results were analyzed using RT‑qPCR and western blot analysis. In the present study, we found that Omi/HtrA2 was overexpressed in hepatocellular carcinoma cell lines and induced hepatocellular carcinoma cell apoptosis. Additiionally, the only manner in which Omi/HtrA2 participated in cell death in PLC cells may be dependent on IAP-binding. Omi/HtrA2‑inducing HepG2 cell apoptosis may mainly depend on its serine protease activity while both IAP-binding and its serine protease activity participated in Hep3B cell apoptosis. This result suggested that Omi/HtrA2 pro-apoptotic marker differs in various hepatocellular carcinoma cell lines. PLC cells were also devoid of the expression of ped/pea-15 as the substrate of Omi/HtrA2 serine protease while ped/pea-15 was overexpressed in HepG2 and Hep3B cells and ped/pea-15 expression was higher in HepG2 cells than that in Hep3B cells. These results showed that Omi/HtrA2 overexpression promotes hepatocellular carcinoma cell apoptosis and the ped/pea-15 expression level causes this difference of the Omi/HtrA2 pro-apoptotic marker in the various hepatocellular carcinoma cell lines.
Collapse
Affiliation(s)
- Zongquan Xu
- Deparment of Hepatic Oncology, Jiangxi Provincial Cancer Hospital, Nanchang 330029, P.R. China
| | - Yu Chen
- Deparment of Hepatic Oncology, Jiangxi Provincial Cancer Hospital, Nanchang 330029, P.R. China
| | - Guohui Xu
- Deparment of Hepatic Oncology, Jiangxi Provincial Cancer Hospital, Nanchang 330029, P.R. China
| | - Cheng Peng
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Enyu Liu
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Yunguang Li
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Jun Niu
- Department of Hepatobiliary Surgery, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Changhai Li
- Hepatic Surgery Center Affiliated Tongji Hospital, Tongji Medical College of HuaZhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
169
|
Perez HL, Chaudhry C, Emanuel SL, Fanslau C, Fargnoli J, Gan J, Kim KS, Lei M, Naglich JG, Traeger SC, Vuppugalla R, Wei DD, Vite GD, Talbott RL, Borzilleri RM. Discovery of Potent Heterodimeric Antagonists of Inhibitor of Apoptosis Proteins (IAPs) with Sustained Antitumor Activity. J Med Chem 2015; 58:1556-62. [DOI: 10.1021/jm501482t] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Heidi L. Perez
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Charu Chaudhry
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Stuart L. Emanuel
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Caroline Fanslau
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joseph Fargnoli
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Jinping Gan
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kyoung S. Kim
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Ming Lei
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joseph G. Naglich
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Sarah C. Traeger
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Ragini Vuppugalla
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Donna D. Wei
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Gregory D. Vite
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Randy L. Talbott
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Robert M. Borzilleri
- Bristol-Myers Squibb Research, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
170
|
Seneci P. Targeting Proteasomal Degradation of Soluble, Misfolded Proteins. CHEMICAL MODULATORS OF PROTEIN MISFOLDING AND NEURODEGENERATIVE DISEASE 2015. [PMCID: PMC7150093 DOI: 10.1016/b978-0-12-801944-3.00003-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This chapter deals with small molecule modulators of the ubiquitin–proteasome system (UPS). They are designed to restore its impaired capacity to dispose of soluble, dysfunctional protein copies, and to fight its pathological impairment in proteinopathies in general and in tauopathies in particular. Two specific molecular targets belonging to the U-box E3 ligase family (C-terminus of Hsc70 interacting protein, CHIP) and to the proteasome-associated cysteine protease DUB family (USP14) are selected for their putative role against NDDs and tauopathies. The limited available structural information for the two targets, and for their interactions with members of UPS-driven protein complexes, is described. A small number of known modulators for each target (or even for structurally related targets, possibly to provide translatable examples) are portrayed in terms of their biological profile, and of their development potential as disease-modifying drugs against NDDs.
Collapse
|
171
|
Li G, Chang H, Zhai YP, Xu W. Targeted silencing of inhibitors of apoptosis proteins with siRNAs: a potential anti-cancer strategy for hepatocellular carcinoma. Asian Pac J Cancer Prev 2014; 14:4943-52. [PMID: 24175757 DOI: 10.7314/apjcp.2013.14.9.4943] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies, with a very poor prognosis. Despite significant improvements in diagnosis and treatment in recent years, the long-term therapeutic efficacy is poor, partially due to tumor metastasis, recurrence, and resistance to chemo- or radio-therapy. Recently, it was found that a major feature of tumors is a combination of unrestrained cell proliferation and impaired apoptosis. There are now 8 recognized members of the IAP-family: NAIP, c-IAP1, c-IAP2, XIAP, Survivin, Bruce, Livin and ILP-2. These proteins all contribute to inhibition of apoptosis, and provide new potential avenues of cancer treatment. As a powerful tool to suppress gene expression in mammalian cells, RNAi species for inhibiting IAP genes can be directed against cancers. This review will provide a brief introduction to recent developments of the application IAP-siRNA in tumor studies, with the aim of inspiring future treatment of HCC.
Collapse
Affiliation(s)
- Gang Li
- Department of General Surgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, China E-mail :
| | | | | | | |
Collapse
|
172
|
Abstract
As the Inhibitor of Apoptosis (IAP) proteins are expressed at high levels in human cancers, they represent promising targets for therapeutic intervention. Small-molecule inhibitors of IAP proteins mimicking the endogenous IAP antagonist Smac, called Smac mimetics, neutralize IAP proteins and thereby promote the induction of cell death. Smac mimetics have been shown in preclinical models of human cancer to directly trigger cancer cell death or to sensitize for cancer cell death induced by a variety of cytotoxic stimuli. Smac mimetics are currently undergoing clinical evaluation in phase I/II trials, demonstrating that therapeutic targeting of IAP proteins has reached the clinical stage.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
173
|
Budhidarmo R, Day CL. IAPs: Modular regulators of cell signalling. Semin Cell Dev Biol 2014; 39:80-90. [PMID: 25542341 DOI: 10.1016/j.semcdb.2014.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/17/2014] [Indexed: 01/30/2023]
Abstract
Members of the inhibitor of apoptosis (IAP) family are characterised by the presence of at least one baculoviral IAP repeat (BIR) domain. However, during the course of evolution, other globular modules have been adopted to perform distinct functions. Consequently, the IAP family is now recognised as consisting of members that perform critical functions in different aspects of cellular regulation. In this review, the structural diversity present within the IAP protein family is presented. Known structures of individual domains are discussed and their properties are described in light of recent data. In particular the plasticity of BIR domains and their ability to accommodate different binding partners is highlighted, as well as the importance of communication between the domains in regulating the covalent attachment of ubiquitin.
Collapse
Affiliation(s)
- Rhesa Budhidarmo
- Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Catherine L Day
- Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
174
|
Profile of Yigong Shi. Proc Natl Acad Sci U S A 2014; 111:16234-5. [DOI: 10.1073/pnas.1418405111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
175
|
Internal motions prime cIAP1 for rapid activation. Nat Struct Mol Biol 2014; 21:1068-74. [DOI: 10.1038/nsmb.2916] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/16/2014] [Indexed: 01/02/2023]
|
176
|
Lee S, Challa-Malladi M, Bratton SB, Wright CW. Nuclear factor-κB-inducing kinase (NIK) contains an amino-terminal inhibitor of apoptosis (IAP)-binding motif (IBM) that potentiates NIK degradation by cellular IAP1 (c-IAP1). J Biol Chem 2014; 289:30680-30689. [PMID: 25246529 PMCID: PMC4215246 DOI: 10.1074/jbc.m114.587808] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/03/2014] [Indexed: 12/20/2022] Open
Abstract
Activation of the noncanonical NF-κB pathway hinges on the stability of the NF-κB-inducing kinase (NIK), which is kept at low levels basally by a protein complex consisting of the E3 ubiquitin ligases cellular inhibitor of apoptosis 1 and 2 (c-IAP1/2) proteins and the tumor necrosis factor receptor-associated factors 2 and 3 (TRAF2/3). NIK is brought into close proximity to the c-IAPs through a TRAF2-TRAF3 bridge where TRAF2 recruits c-IAP1/2 and TRAF3 binds to NIK. However, it is not clear how the c-IAPs specifically recognize and ubiquitylate NIK in the complex. We have identified an IAP-binding motif (IBM) at the amino terminus of NIK. IBMs are utilized by a number of proapoptotic proteins to antagonize IAP function. Here, we utilize mutational studies to demonstrate that wild-type NIK is destabilized in the presence of c-IAP1, whereas the NIK IBM mutant is stable. NIK interacts with the second baculovirus IAP repeat (BIR2) domain of c-IAP1 via the IBM, and this interaction, in turn, provides substrate recognition for c-IAP1 mediated ubiquitylation and degradation of NIK. Furthermore, in the presence of the NIK IBM mutant, we observed an elevated processing of p100 to p52 followed by increased expression of NF-κB target genes. Together, these findings reveal the novel identification and function of the NIK IBM, which promotes c-IAP1-dependent ubiquitylation of NIK, resulting in optimal NIK turnover to ensure that noncanonical NF-κB signaling is off in the absence of an activating signal.
Collapse
Affiliation(s)
- Sunhee Lee
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, and
| | - Madhavi Challa-Malladi
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, and
| | - Shawn B Bratton
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, and; Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park, Smithville, Texas 78957
| | - Casey W Wright
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, and; Center for Molecular and Cellular Toxicology in the Division of Pharmacology and Toxicology, College of Pharmacy and The University of Texas at Austin, Austin, Texas 78712.
| |
Collapse
|
177
|
PARK BYOUNGDUCK. JNK1-mediated phosphorylation of Smac/DIABLO at the serine 6 residue is functionally linked to its mitochondrial release during TNF-α-induced apoptosis of HeLa cells. Mol Med Rep 2014; 10:3205-10. [DOI: 10.3892/mmr.2014.2625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 06/20/2014] [Indexed: 11/05/2022] Open
|
178
|
Elkholi R, Renault TT, Serasinghe MN, Chipuk JE. Putting the pieces together: How is the mitochondrial pathway of apoptosis regulated in cancer and chemotherapy? Cancer Metab 2014; 2:16. [PMID: 25621172 PMCID: PMC4304082 DOI: 10.1186/2049-3002-2-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/20/2014] [Indexed: 02/08/2023] Open
Abstract
In order to solve a jigsaw puzzle, one must first have the complete picture to logically connect the pieces. However, in cancer biology, we are still gaining an understanding of all the signaling pathways that promote tumorigenesis and how these pathways can be pharmacologically manipulated by conventional and targeted therapies. Despite not having complete knowledge of the mechanisms that cause cancer, the signaling networks responsible for cancer are becoming clearer, and this information is serving as a solid foundation for the development of rationally designed therapies. One goal of chemotherapy is to induce cancer cell death through the mitochondrial pathway of apoptosis. Within this review, we present the pathways that govern the cellular decision to undergo apoptosis as three distinct, yet connected puzzle pieces: (1) How do oncogene and tumor suppressor pathways regulate apoptosis upstream of mitochondria? (2) How does the B-cell lymphoma 2 (BCL-2) family influence tumorigenesis and chemotherapeutic responses? (3) How is post-mitochondrial outer membrane permeabilization (MOMP) regulation of cell death relevant in cancer? When these pieces are united, it is possible to appreciate how cancer signaling directly impacts upon the fundamental cellular mechanisms of apoptosis and potentially reveals novel pharmacological targets within these pathways that may enhance chemotherapeutic success.
Collapse
Affiliation(s)
- Rana Elkholi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1425 Madison Avenue, Box 1130, New York, NY 10029, USA.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA
| | - Thibaud T Renault
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1425 Madison Avenue, Box 1130, New York, NY 10029, USA.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA
| | - Madhavika N Serasinghe
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1425 Madison Avenue, Box 1130, New York, NY 10029, USA.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1425 Madison Avenue, Box 1130, New York, NY 10029, USA.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.,The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA
| |
Collapse
|
179
|
Zhang J, Zhong Q. Histone deacetylase inhibitors and cell death. Cell Mol Life Sci 2014; 71:3885-901. [PMID: 24898083 PMCID: PMC4414051 DOI: 10.1007/s00018-014-1656-6] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/23/2014] [Accepted: 05/20/2014] [Indexed: 12/14/2022]
Abstract
Histone deacetylases (HDACs) are a vast family of enzymes involved in chromatin remodeling and have crucial roles in numerous biological processes, largely through their repressive influence on transcription. In addition to modifying histones, HDACs also target many other non-histone protein substrates to regulate gene expression. Recently, HDACs have gained growing attention as HDAC-inhibiting compounds are being developed as promising cancer therapeutics. Histone deacetylase inhibitors (HDACi) have been shown to induce differentiation, cell cycle arrest, apoptosis, autophagy and necrosis in a variety of transformed cell lines. In this review, we mainly discuss how HDACi may elicit a therapeutic response to human cancers through different cell death pathways, in particular, apoptosis and autophagy.
Collapse
Affiliation(s)
- Jing Zhang
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
180
|
Bai L, Smith DC, Wang S. Small-molecule SMAC mimetics as new cancer therapeutics. Pharmacol Ther 2014; 144:82-95. [PMID: 24841289 PMCID: PMC4247261 DOI: 10.1016/j.pharmthera.2014.05.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/19/2022]
Abstract
Apoptosis is a tightly regulated cellular process and faulty regulation of apoptosis is a hallmark of human cancers. Targeting key apoptosis regulators with the goal to restore apoptosis in tumor cells has been pursued as a new cancer therapeutic strategy. XIAP, cIAP1, and cIAP2, members of inhibitor of apoptosis (IAP) proteins, are critical regulators of cell death and survival and are attractive targets for new cancer therapy. The SMAC/DIABLO protein is an endogenous antagonist of XIAP, cIAP1, and cIAP2. In the last decade, intense research efforts have resulted in the design and development of several small-molecule SMAC mimetics now in clinical trials for cancer treatment. In this review, we will discuss the roles of XIAP, cIAP1, and cIAP2 in regulation of cell death and survival, and the design and development of small-molecule SMAC mimetics as novel cancer treatments.
Collapse
Affiliation(s)
- Longchuan Bai
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - David C Smith
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| | - Shaomeng Wang
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
181
|
Pieczykolan JS, Kubiński K, Masłyk M, Pawlak SD, Pieczykolan A, Rózga PK, Szymanik M, Gałązka M, Teska-Kamińska M, Żerek B, Bukato K, Poleszak K, Jaworski A, Strożek W, Świder R, Zieliński R. AD-O53.2--a novel recombinant fusion protein combining the activities of TRAIL/Apo2L and Smac/Diablo, overcomes resistance of human cancer cells to TRAIL/Apo2L. Invest New Drugs 2014; 32:1155-66. [PMID: 25182378 DOI: 10.1007/s10637-014-0153-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/26/2014] [Indexed: 10/24/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptors became promising molecules for selective targeting of tumor cells without affecting normal tissue. Unfortunately, cancer cells have developed a number of mechanisms that confer resistance to TRAIL\Apo2L-induced apoptosis, which substantiates the need for development of alternative therapeutic strategies. Here we present a recombinant variant of TRAIL\Apo2L peptide, named AD-O53.2, fused to the peptide-derived from Smac/Diablo protein-the natural inhibitor of the apoptotic X-linked IAP (XIAP) protein considered as a pro-apoptotic agent. The proposed mechanism of action for this construct involves specific targeting of the tumor by TRAIL\Apo2L followed by activation and internalization of pro-apoptotic peptide into the cancer cells. While in the cytoplasm , the Smac\Diablo peptide inhibits activity of X-linked IAP (XIAP) proteins and promotes caspase-mediated apoptosis. AD-O53.2 construct was expressed in E.coli and purified by Ion Exchange Chromatography (IEC). Derived protein was initially characterized by circular dichroism spectroscopy (CD), HPLC-SEC chromatography, surface plasmon resonance, protease activation and cell proliferation assays. Our Smac/Diablo-TRAIL fusion variant was tested against a panel of cancer cells (including lung, colorectal, pancreatic, liver, kidney and uterine) and showed a potent cytotoxic effect with the IC50 values in femtomolar range for the most sensitive cell lines, while it remained ineffective against non-transformed HUVEC cells as well as isolated normal human and rat hepatocytes. Importantly, the construct was well tolerated by animals and significantly reduced the rate of the tumor growth in colon and lung adenocarcinoma animal models.
Collapse
Affiliation(s)
- Jerzy S Pieczykolan
- Drug Discovery Department, Adamed Group, Pieńków 149, 05-152, Czosnów, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Bourguet CB, Boulay PL, Claing A, Lubell WD. Design and synthesis of novel azapeptide activators of apoptosis mediated by caspase-9 in cancer cells. Bioorg Med Chem Lett 2014; 24:3361-5. [DOI: 10.1016/j.bmcl.2014.05.095] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 05/26/2014] [Accepted: 05/28/2014] [Indexed: 10/25/2022]
|
183
|
Li CF, Chen LB, Li DD, Yang L, Zhang BG, Jin JP, Zhang Y, Zhang B. Dual‑sensitive HRE/Egr1 promoter regulates Smac overexpression and enhances radiation‑induced A549 human lung adenocarcinoma cell death under hypoxia. Mol Med Rep 2014; 10:1108-1116. [PMID: 24842518 DOI: 10.3892/mmr.2014.2233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 04/04/2014] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to construct an expression vector carrying the hypoxia/radiation dual‑sensitive chimeric hypoxia response element (HRE)/early growth response 1 (Egr‑1) promoter in order to overexpress the therapeutic second mitochondria‑derived activator of caspases (Smac). Using this expression vector, the present study aimed to explore the molecular mechanism underlying radiotherapy‑induced A549 human lung adenocarcinoma cell death and apoptosis under hypoxia. The plasmids, pcDNA3.1‑Egr1‑Smac (pE‑Smac) and pcDNA3.1‑HRE/Egr-1‑Smac (pH/E‑Smac), were constructed and transfected into A549 human lung adenocarcinoma cells using the liposome method. CoCl2 was used to chemically simulate hypoxia, followed by the administration of 2 Gy X‑ray irradiation. An MTT assay was performed to detect cell proliferation and an Annexin V‑fluorescein isothiocyanate apoptosis detection kit was used to detect apoptosis. Quantitative polymerase chain reaction and western blot analyses were used for the detection of mRNA and protein expression, respectively. Infection with the pE‑Smac and pH/E‑Smac plasmids in combination with radiation and/or hypoxia was observed to enhance the expression of Smac. Furthermore, Smac overexpression was found to enhance the radiation‑induced inhibition of cell proliferation and promotion of cycle arrest and apoptosis. The cytochrome c/caspase‑9/caspase‑3 pathway was identified to be involved in this regulation of apoptosis. Plasmid infection in combination with X‑ray irradiation was found to markedly induce cell death under hypoxia. In conclusion, the hypoxia/radiation dual‑sensitive chimeric HRE/Egr‑1 promoter was observed to enhance the expression of the therapeutic Smac, as well as enhance the radiation‑induced inhibition of cell proliferation and promotion of cycle arrest and apoptosis under hypoxia. This apoptosis was found to involve the mitochondrial pathway.
Collapse
Affiliation(s)
- Chang-Feng Li
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Li-Bo Chen
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Dan-Dan Li
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Lei Yang
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Bao-Gang Zhang
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Jing-Peng Jin
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ying Zhang
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Bin Zhang
- Endoscopy Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
184
|
Xiong S, Mu T, Wang G, Jiang X. Mitochondria-mediated apoptosis in mammals. Protein Cell 2014; 5:737-49. [PMID: 25073422 PMCID: PMC4180462 DOI: 10.1007/s13238-014-0089-1] [Citation(s) in RCA: 313] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/08/2014] [Indexed: 01/06/2023] Open
Abstract
The mitochondria-mediated caspase activation pathway is a major apoptotic pathway characterized by mitochondrial outer membrane permeabilization (MOMP) and subsequent release of cytochrome c into the cytoplasm to activate caspases. MOMP is regulated by the Bcl-2 family of proteins. This pathway plays important roles not only in normal development, maintenance of tissue homeostasis and the regulation of immune system, but also in human diseases such as immune disorders, neurodegeneration and cancer. In the past decades the molecular basis of this pathway and the regulatory mechanism have been comprehensively studied, yet a great deal of new evidence indicates that cytochrome c release from mitochondria does not always lead to irreversible cell death, and that caspase activation can also have non-death functions. Thus, many unsolved questions and new challenges are still remaining. Furthermore, the dysfunction of this pathway involved in cancer development is obvious, and targeting the pathway as a therapeutic strategy has been extensively explored, but the efficacy of the targeted therapies is still under development. In this review we will discuss the mitochondria-mediated apoptosis pathway and its physiological roles and therapeutic implications.
Collapse
Affiliation(s)
- Shunbin Xiong
- Department of Genetics, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | | | | | | |
Collapse
|
185
|
Barile E, Pellecchia M. NMR-based approaches for the identification and optimization of inhibitors of protein-protein interactions. Chem Rev 2014; 114:4749-63. [PMID: 24712885 PMCID: PMC4027952 DOI: 10.1021/cr500043b] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Elisa Barile
- Sanford-Burnham Medical
Research Institute, 10901
North Torrey Pines Road, La Jolla, California 92037, United States
| | - Maurizio Pellecchia
- Sanford-Burnham Medical
Research Institute, 10901
North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
186
|
Wang X, Piro B, Reisberg S, Anquetin G, de Rocquigny H, Jiang P, Wang Q, Wu W, Pham MC, Dong CZ. Direct, reagentless electrochemical detection of the BIR3 domain of X-linked inhibitor of apoptosis protein using a peptide-based conducting polymer sensor. Biosens Bioelectron 2014; 61:57-62. [PMID: 24858673 DOI: 10.1016/j.bios.2014.04.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/09/2014] [Accepted: 04/23/2014] [Indexed: 10/25/2022]
Abstract
In this work, we report a reagentless electrochemical peptide (AVPFAQKG) sensor to directly detect the BIR3 domain of X-linked inhibitor of apoptosis protein (XIAP-BIR3). The bioreceptor was based on a conducting copolymer film electrosynthesized from juglone and a juglone-peptide conjugate (JP) newly designed. The peptide-protein interactions generated an important increase of steric hindrance at the interface and a current decrease (signal off) of the redox reaction from quinone embedded in the polymer backbone as evidenced by Square Wave Voltammetry. This allowed a specific and sensitive detection of XIAP-BIR3 with a detection limit of 1 nM (13 ng mL(-1)). The peptide-protein complex could be then dissociated by adding the free precursor peptide (AVPFAQKG) into solution, causing a shift-back on the signal, i.e. an increase in the current intensity (signal-on). This "off-on" detection sequence was used in this work as a double verification of the specificity and this approach can be employed as a general way to increase the reliability of the results. In general, the approach described in this work may be inspired to develop other direct and reagentless electrochemical protein assays with high specificity and sensitivity.
Collapse
Affiliation(s)
- X Wang
- Lanzhou University, School of Nuclear Science and Technology, Lanzhou 730000, China; University Paris Diderot, Sorbonne Paris Cité, ITODYS, UMR CNRS 7086, 15 rue J-A de Baïf, 75205 Paris Cedex 13, France
| | - B Piro
- University Paris Diderot, Sorbonne Paris Cité, ITODYS, UMR CNRS 7086, 15 rue J-A de Baïf, 75205 Paris Cedex 13, France.
| | - S Reisberg
- University Paris Diderot, Sorbonne Paris Cité, ITODYS, UMR CNRS 7086, 15 rue J-A de Baïf, 75205 Paris Cedex 13, France
| | - G Anquetin
- University Paris Diderot, Sorbonne Paris Cité, ITODYS, UMR CNRS 7086, 15 rue J-A de Baïf, 75205 Paris Cedex 13, France
| | - H de Rocquigny
- Université de Strasbourg, UMR CNRS 7213, Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, 67401 Illkirch, France
| | - P Jiang
- Lanzhou University, Institute of Cancer Biology and Drug Screening, School of Life Sciences, Lanzhou 730000, China
| | - Q Wang
- Lanzhou University, Institute of Cancer Biology and Drug Screening, School of Life Sciences, Lanzhou 730000, China
| | - W Wu
- Lanzhou University, School of Nuclear Science and Technology, Lanzhou 730000, China
| | - M-C Pham
- University Paris Diderot, Sorbonne Paris Cité, ITODYS, UMR CNRS 7086, 15 rue J-A de Baïf, 75205 Paris Cedex 13, France
| | - C-Z Dong
- University Paris Diderot, Sorbonne Paris Cité, ITODYS, UMR CNRS 7086, 15 rue J-A de Baïf, 75205 Paris Cedex 13, France; Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan 430056, China.
| |
Collapse
|
187
|
Wang J, Li W. Discovery of novel second mitochondria-derived activator of caspase mimetics as selective inhibitor of apoptosis protein inhibitors. J Pharmacol Exp Ther 2014; 349:319-29. [PMID: 24623800 PMCID: PMC3989805 DOI: 10.1124/jpet.113.212019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/11/2014] [Indexed: 01/19/2023] Open
Abstract
Inhibitor of apoptosis (IAP) proteins are widely considered as promising cancer drug targets, especially for drug-resistant tumors. Mimicking the IAP-binding motif of second mitochondria-derived activator of caspases (SMAC) is a rational strategy to design potential IAP inhibitors. In this report, we used the bioactive conformation of AVPI tetrapeptide in the N terminus of SMAC as a template and performed a shape-based virtual screening against a drug-like compound library to identify novel IAP inhibitors. Top hits were subsequently docked to available IAP crystal structures as a secondary screening followed by validation using in vitro biologic assays. Four novel hit compounds were identified to potently inhibit cell growth in two human melanoma (A375 and M14) and two human prostate (PC-3 and DU145) cancer cell lines. The best compound, UC-112 [5-((benzyloxy)methyl)-7-(pyrrolidin-1-ylmethyl)quinolin-8-ol], has IC50 values ranging from 0.7 to 3.4 µM. UC-112 also potently inhibits the growth of P-glycoprotein (P-gp)-overexpressed multidrug-resistant cancer cells, strongly activates caspase-3/7 and caspase-9 activities, and selectively downregulates survivin level at a concentration as low as 1 µM. Coincubation of UC-112 with a known proteasome inhibitor Z-Leu-Leu-Leu-CHO (MG-132) rescued survivin inhibition, consistent with the anticipated mechanism of action for UC-112. As a single agent, UC-112 strongly inhibits tumor growth and reduces both X chromosome-linked IAP and survivin levels in an A375 human melanoma xenograft model in vivo. Overall, our study identified novel scaffolds, especially UC-112, as new platforms on which potent and selective IAP antagonists can be developed.
Collapse
Affiliation(s)
- Jin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee
| | | |
Collapse
|
188
|
Sun H, Lu J, Liu L, Yang CY, Wang S. Potent and selective small-molecule inhibitors of cIAP1/2 proteins reveal that the binding of Smac mimetics to XIAP BIR3 is not required for their effective induction of cell death in tumor cells. ACS Chem Biol 2014; 9:994-1002. [PMID: 24521431 PMCID: PMC4324444 DOI: 10.1021/cb400889a] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Cellular
inhibitor of apoptosis protein 1 and 2 (cIAP1/2) and X-linked
inhibitor of apoptosis protein (XIAP) are key apoptosis regulators
and promising new cancer therapeutic targets. This study describes
a set of non-peptide, small-molecule Smac (second mitochondria-derived
activator of caspases) mimetics that are selective inhibitors of cIAP1/2
over XIAP. The most potent and most selective compounds bind to cIAP1/2
with affinities in the low nanomolar range and show >1,000-fold
selectivity
for cIAP1 over XIAP. These selective cIAP inhibitors effectively induce
degradation of the cIAP1 protein in cancer cells at low nanomolar
concentrations and do not antagonize XIAP in a cell-free functional
assay. They potently inhibit cell growth and effectively induce apoptosis
at low nanomolar concentrations in cancer cells with a mechanism of
action similar to that of other known Smac mimetics. Our study shows
that binding of Smac mimetics to XIAP BIR3 is not required for effective
induction of apoptosis in tumor cells by Smac mimetics. These potent
and highly selective cIAP1/2 inhibitors are powerful tools in the
investigation of the role of these IAP proteins in the regulation
of apoptosis and other cellular processes.
Collapse
Affiliation(s)
- Haiying Sun
- Comprehensive Cancer Center
and Departments of Internal Medicine, Pharmacology, and Medicinal
Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Jianfeng Lu
- Comprehensive Cancer Center
and Departments of Internal Medicine, Pharmacology, and Medicinal
Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Liu Liu
- Comprehensive Cancer Center
and Departments of Internal Medicine, Pharmacology, and Medicinal
Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Chao-Yie Yang
- Comprehensive Cancer Center
and Departments of Internal Medicine, Pharmacology, and Medicinal
Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Comprehensive Cancer Center
and Departments of Internal Medicine, Pharmacology, and Medicinal
Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
189
|
Condon SM, Mitsuuchi Y, Deng Y, LaPorte MG, Rippin SR, Haimowitz T, Alexander MD, Kumar PT, Hendi MS, Lee YH, Benetatos CA, Yu G, Kapoor GS, Neiman E, Seipel ME, Burns JM, Graham MA, McKinlay MA, Li X, Wang J, Shi Y, Feltham R, Bettjeman B, Cumming MH, Vince JE, Khan N, Silke J, Day CL, Chunduru SK. Birinapant, a smac-mimetic with improved tolerability for the treatment of solid tumors and hematological malignancies. J Med Chem 2014; 57:3666-77. [PMID: 24684347 DOI: 10.1021/jm500176w] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Birinapant (1) is a second-generation bivalent antagonist of IAP proteins that is currently undergoing clinical development for the treatment of cancer. Using a range of assays that evaluated cIAP1 stability and oligomeric state, we demonstrated that 1 stabilized the cIAP1-BUCR (BIR3-UBA-CARD-RING) dimer and promoted autoubiquitylation of cIAP1 in vitro. Smac-mimetic 1-induced loss of cIAPs correlated with inhibition of TNF-mediated NF-κB activation, caspase activation, and tumor cell killing. Many first-generation Smac-mimetics such as compound A (2) were poorly tolerated. Notably, animals that lack functional cIAP1, cIAP2, and XIAP are not viable, and 2 mimicked features of triple IAP knockout cells in vitro. The improved tolerability of 1 was associated with (i) decreased potency against cIAP2 and affinity for XIAP BIR3 and (ii) decreased ability to inhibit XIAP-dependent signaling pathways. The P2' position of 1 was critical to this differential activity, and this improved tolerability has allowed 1 to proceed into clinical studies.
Collapse
Affiliation(s)
- Stephen M Condon
- TetraLogic Pharmaceuticals, Inc. , 343 Phoenixville Pike, Malvern, Pennsylvania 19355, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Carter BZ, Mak PY, Mak DH, Shi Y, Qiu Y, Bogenberger JM, Mu H, Tibes R, Yao H, Coombes KR, Jacamo RO, McQueen T, Kornblau SM, Andreeff M. Synergistic targeting of AML stem/progenitor cells with IAP antagonist birinapant and demethylating agents. J Natl Cancer Inst 2014; 106:djt440. [PMID: 24526787 DOI: 10.1093/jnci/djt440] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) therapy has limited long-term efficacy because patients frequently develop disease relapse because of the inability of standard chemotherapeutic agents to target AML stem/progenitor cells. Here, we identify deregulated apoptotic components in AML stem/progenitor cells and investigate the individual and combinatorial effects of the novel inhibitor of apoptosis (IAP) protein antagonist and second mitochondrial-derived activator of caspases (SMAC) mimetic birinapant and demethylating epigenetic modulators. METHODS Protein expression was measured by reversed-phase protein array in AML patient (n = 511) and normal (n = 21) samples and by western blot in drug-treated cells. The antileukemic activity of birinapant and demethylating agents was assessed in vitro and in an in vivo AML mouse xenograft model (n = 10 mice per group). All statistical tests were two-sided. RESULTS Compared with bulk AML cells, CD34(+)38(-) AML stem/progenitors expressed increased cIAP1 and caspase-8 levels and decreased SMAC levels (one-way analysis of variance followed by Tukey's multiple comparison test, P < .001). Birinapant induced death receptor-/caspase-8-mediated apoptosis in AML cells, including in AML stem/progenitor cells, but not in normal CD34(+) cells. Demethylating agents modulated extrinsic apoptosis pathway components and, when combined with birinapant, were highly synergistic in vitro (combination index < 1), and also more effective in vivo (P < .001, by Student t test, for the median survival of birinapant plus 5-azacytadine vs birinapant alone or vs controls). CONCLUSIONS cIAP1, SMAC, and caspase-8 appear to play a role in AML stem cell survival, and synergistic targeting of these cells with birinapant and demethylating agents shows potential utility in leukemia therapy.
Collapse
Affiliation(s)
- Bing Z Carter
- Affiliations of authors: Section of Molecular Hematology and Therapy, Department of Leukemia (BZC, PYM, DHM, YS, YQ, HM, ROJ, TM, SMK, MA) and Department of Bioinformatics and Computational Biology (HY, KRC), University of Texas M.D. Anderson Cancer Center, Houston, TX; Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ (JMB, RT)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Aeluri M, Chamakuri S, Dasari B, Guduru SKR, Jimmidi R, Jogula S, Arya P. Small Molecule Modulators of Protein–Protein Interactions: Selected Case Studies. Chem Rev 2014; 114:4640-94. [DOI: 10.1021/cr4004049] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Madhu Aeluri
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Srinivas Chamakuri
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Bhanudas Dasari
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Shiva Krishna Reddy Guduru
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Ravikumar Jimmidi
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Srinivas Jogula
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| | - Prabhat Arya
- Dr. Reddy’s Institute
of Life Sciences (DRILS), University of Hyderabad Campus Gachibowli, Hyderabad 500046, India
| |
Collapse
|
192
|
Wehrkamp CJ, Gutwein AR, Natarajan SK, Phillippi MA, Mott JL. XIAP antagonist embelin inhibited proliferation of cholangiocarcinoma cells. PLoS One 2014; 9:e90238. [PMID: 24603802 PMCID: PMC3946004 DOI: 10.1371/journal.pone.0090238] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/26/2014] [Indexed: 01/07/2023] Open
Abstract
Cholangiocarcinoma cells are dependent on antiapoptotic signaling for survival and resistance to death stimuli. Recent mechanistic studies have revealed that increased cellular expression of the E3 ubiquitin-protein ligase X-linked inhibitor of apoptosis (XIAP) impairs TRAIL- and chemotherapy-induced cytotoxicity, promoting survival of cholangiocarcinoma cells. This study was undertaken to determine if pharmacologic antagonism of XIAP protein was sufficient to sensitize cholangiocarcinoma cells to cell death. We employed malignant cholangiocarcinoma cell lines and used embelin to antagonize XIAP protein. Embelin treatment resulted in decreased XIAP protein levels by 8 hours of treatment with maximal effect at 16 hours in KMCH and Mz-ChA-1 cells. Assessment of nuclear morphology demonstrated a concentration-dependent increase in nuclear staining. Interestingly, embelin induced nuclear morphology changes as a single agent, independent of the addition of TNF-related apoptosis inducing ligand (TRAIL). However, caspase activity assays revealed that increasing embelin concentrations resulted in slight inhibition of caspase activity, not activation. In addition, the use of a pan-caspase inhibitor did not prevent nuclear morphology changes. Finally, embelin treatment of cholangiocarcinoma cells did not induce DNA fragmentation or PARP cleavage. Apoptosis does not appear to contribute to the effects of embelin on cholangiocarcinoma cells. Instead, embelin caused inhibition of cell proliferation and cell cycle analysis indicated that embelin increased the number of cells in S and G2/M phase. Our results demonstrate that embelin decreased proliferation in cholangiocarcinoma cell lines. Embelin treatment resulted in decreased XIAP protein expression, but did not induce or enhance apoptosis. Thus, in cholangiocarcinoma cells the mechanism of action of embelin may not be dependent on apoptosis.
Collapse
Affiliation(s)
- Cody J. Wehrkamp
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ashley R. Gutwein
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sathish Kumar Natarajan
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Mary Anne Phillippi
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
193
|
Huber HJ, McKiernan RG, Prehn JHM. Harnessing system models of cell death signalling for cytotoxic chemotherapy: towards personalised medicine approaches? J Mol Med (Berl) 2014; 92:227-37. [PMID: 24477766 DOI: 10.1007/s00109-014-1126-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/09/2014] [Accepted: 01/14/2014] [Indexed: 12/27/2022]
Abstract
Most cytotoxic chemotherapeutics are believed to kill cancer cells by inducing apoptosis. Understanding the factors that contribute to impairment of apoptosis in cancer cells is therefore critical for the development of novel therapies that circumvent the widespread chemoresistance. Apoptosis, however, is a complex and tightly controlled process that can be induced by different classes of chemotherapeutics targeting different signalling nodes and pathways. Moreover, apoptosis initiation and apoptosis execution strongly depend on patient-specific, genomic and proteomic signatures. Here, we will review recent translational studies that suggest a critical link between the sensitivity of cancer cells to initiate apoptosis and clinical outcome. Next we will discuss recent advances in the field of system modelling of apoptosis pathways for the prediction of treatment responses. We propose that initiation of mitochondrial apoptosis, defined as the process of mitochondrial outer membrane permeabilisation (MOMP), is a dose-dependent decision process that allows for a prediction of individual therapy responses and therapeutic windows. We provide evidence in contrast that apoptosis execution post-MOMP may be a binary decision that dictates whether apoptosis is executed or not. We will discuss the implications of this concept for the future use of novel adjuvant therapeutics that specifically target apoptosis signalling pathways or which may be used to reduce the impact of cell-to-cell heterogeneity on therapy responses. Finally, we will discuss the technical and regulatory requirements surrounding the use and implications of system-based patient stratification tools for the future of personalised oncology.
Collapse
Affiliation(s)
- Heinrich J Huber
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland,
| | | | | |
Collapse
|
194
|
Micewicz ED, Luong HT, Jung CL, Waring AJ, McBride WH, Ruchala P. Novel dimeric Smac analogs as prospective anticancer agents. Bioorg Med Chem Lett 2014; 24:1452-7. [PMID: 24582479 DOI: 10.1016/j.bmcl.2014.02.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/05/2014] [Accepted: 02/07/2014] [Indexed: 12/20/2022]
Abstract
A small library of monovalent Smac mimics with general structure NMeAla-Tle-(4R)-4-Benzyl-Pro-Xaa-cysteamide, was synthesized (Xaa=hydrophobic residue). The library was screened in vitro against human breast cancer cell lines MCF-7 and MDA-MB-231, and two most active compounds oligomerized via S-alkylation giving bivalent and trivalent derivatives. The most active bivalent analogue SMAC17-2X was tested in vivo and in physiological conditions (mouse model) it exerted a potent anticancer effect resulting in ∼23.4days of tumor growth delay at 7.5mg/kg dose. Collectively, our findings suggest that bivalent Smac analogs obtained via S-alkylation protocol may be a suitable platform for the development of new anticancer therapeutics.
Collapse
Affiliation(s)
- Ewa D Micewicz
- Department of Radiation Oncology, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Hai T Luong
- Department of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Chun-Ling Jung
- Department of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Alan J Waring
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90502, USA; Department of Physiology and Biophysics, University of California Irvine, 1001 Health Sciences Road, Irvine, CA 92697, USA
| | - William H McBride
- Department of Radiation Oncology, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Piotr Ruchala
- Department of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA.
| |
Collapse
|
195
|
Survivin as a preferential target for cancer therapy. Int J Mol Sci 2014; 15:2494-516. [PMID: 24531137 PMCID: PMC3958864 DOI: 10.3390/ijms15022494] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 01/31/2014] [Accepted: 02/07/2014] [Indexed: 11/16/2022] Open
Abstract
Cancer is typically a consequence of imbalance between cell death and proliferation in a way favorable to cell proliferation and survival. Most conventional cancer therapies are based on targeting rapidly growing cancerous cells to block growth or enhance cell death, thereby, restoring the balance between these processes. In many instances, malignancies that develop resistance to current treatment modalities, such as chemotherapy, immunotherapy, and radiotherapy often present the greatest challenge in subsequent management of the patient. Studies have shown that under normal circumstances, cells utilize different death mechanisms, such as apoptosis (programmed cell death), autophagy, mitotic catastrophe, and necrosis to maintain homeostasis and physiological integrity of the organism, but these processes often appear to be altered in cancer. Thus, in recent years developing various strategies for administration of cytotoxic chemotherapeutics in combination with apoptosis-sensitizing reagents is receiving more emphasis. Here, we review the properties of the anti-apoptotic protein, survivin, a member of the inhibitor of apoptosis protein (IAP) family and the clinical feasibility and anti-cancer potential of drugs targeting this protein. We also discuss some key points and concerns that should be taken into consideration while developing drugs that target apoptotic proteins, such as survivin.
Collapse
|
196
|
Abstract
Apoptosis is a cell death program that is well-orchestrated for normal tissue homeostasis and for removal of damaged, old or infected cells. It is regulated by intrinsic and extrinsic pathways. The intrinsic pathway responds to signals such as ultraviolet radiation or DNA damage and activates "executioner" caspases through a mitochondria-dependent pathway. The extrinsic pathway is activated by death signals induced, for example, by an infection that activates the immune system or receptor-mediated pathways. The extrinsic pathway signals also cascade down to executioner caspases that cleave target proteins and lead to cell death. Strict control of cellular apoptosis is important for the hematopoietic system as it has a high turnover rate. However, the apoptosis program is often deregulated in hematologic malignancies leading to the accumulation of malignant cells. Therefore, apoptosis pathways have been identified for the development of anticancer therapeutics. We review here the proteins that have been targeted for anticancer drug development in hematologic malignancies. These include BCL-2 family proteins, death ligands and receptors, inhibitor of apoptosis family proteins and caspases. Except for caspase activators, drugs that target each of these classes of proteins have advanced into clinical trials.
Collapse
Affiliation(s)
- Shadia Zaman
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center , Houston, TX , USA
| | | | | |
Collapse
|
197
|
Abstract
Cell death by apoptosis plays a critical role in regulating the subtle balance between cell death and proliferation to maintain tissue homeostasis. Accordingly, tipping the balance in either direction may cause human disease. Too little cell death may promote tumor formation and progression. In addition, killing of cancer cells by current therapies is largely due to induction of apoptosis in tumor cells. Since a hallmark of human cancers is their resistance to apoptosis, there is a demand to develop novel strategies that restore the apoptotic machinery in order to overcome cancer resistance. Inhibitor of apoptosis proteins (IAPs) block apoptosis at the core of the apoptotic machinery by inhibiting caspases. Elevated levels of IAPs are found in many human cancers and have been associated with poor prognosis. Recent insights into the role of IAPs have provided the basis for various exciting developments that aim to modulate the expression or function of IAPs in human cancers. Targeting IAPs (e.g., by antisense approaches or small-molecule inhibitors) presents a promising novel approach to either directly trigger apoptosis or to potentiate the efficacy of cytotoxic therapies in cancer cells. Thus, inhibition of IAPs such as X chromosome-linked IAP may prove to be a successful strategy to overcome apoptosis resistance of human cancers that deserves further exploitation.
Collapse
Affiliation(s)
- Simone Fulda
- University Children's Hospital, Eythstr. 24-89075, Ulm, Germany.
| |
Collapse
|
198
|
Inhibitor of apoptosis proteins as therapeutic targets in multiple myeloma. Leukemia 2014; 28:1519-28. [PMID: 24402161 PMCID: PMC4090267 DOI: 10.1038/leu.2014.2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 12/20/2013] [Indexed: 12/26/2022]
Abstract
The inhibitor of apoptosis (IAP) proteins plays a critical role in the control of apoptotic machinery, and has been explored as a therapeutic target. Here, we have examined the functional importance of IAPs in multiple myeloma (MM) by using a Smac-mimetic LCL161. We observed that LCL161 was able to potently induce apoptosis in some MM cell lines but not in others. Examining the levels of XIAP, cIAP1 and cIAP2 post LCL161 treatment indicated clear down regulation of both XIAP activity and cIAP1 levels in both the sensitive and less sensitive (resistant) cell lines. cIAP2, however, was not down regulated in the cell line resistant to the drug. siRNA mediated silencing of cIAP2 significantly enhanced the effect of LCL161 indicating the importance of down regulating all IAPs simultaneously for induction of apopotsis in MM cells. LCL161 induced marked up regulation of the Jak2/Stat3 pathway in the resistant MM cell lines. Combining LCL161 with a Jak2 specific inhibitor resulted in synergistic cell death in MM cell lines and patient cells. In addition, combining LCL161 with death inducing ligands clearly showed that LCL161 sensitized MM cells to both FAS-L and TRAIL.
Collapse
|
199
|
Yuan Z, Pastoriza J, Quinn T, Libutti SK. Targeting Tumor Vasculature Using Adeno-Associated Virus Phage Vectors Coding Tumor Necrosis Factor-α. GENE THERAPY OF CANCER 2014:19-33. [DOI: 10.1016/b978-0-12-394295-1.00002-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
200
|
Kleino A, Silverman N. The Drosophila IMD pathway in the activation of the humoral immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 42:25-35. [PMID: 23721820 PMCID: PMC3808521 DOI: 10.1016/j.dci.2013.05.014] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 05/17/2013] [Accepted: 05/17/2013] [Indexed: 05/08/2023]
Abstract
The IMD pathway signaling plays a pivotal role in the Drosophila defense against bacteria. During the last two decades, significant progress has been made in identifying the components and deciphering the molecular mechanisms underlying this pathway, including the means of bacterial sensing and signal transduction. While these findings have contributed to the understanding of the immune signaling in insects, they have also provided new insights in studying the mammalian NF-κB signaling pathways. Here, we summarize the current view of the IMD pathway focusing on how it regulates the humoral immune response of Drosophila.
Collapse
Affiliation(s)
- Anni Kleino
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | |
Collapse
|