51
|
Kocik J, Machula M, Wisniewska A, Surmiak E, Holak TA, Skalniak L. Helping the Released Guardian: Drug Combinations for Supporting the Anticancer Activity of HDM2 (MDM2) Antagonists. Cancers (Basel) 2019; 11:E1014. [PMID: 31331108 PMCID: PMC6678622 DOI: 10.3390/cancers11071014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 01/22/2023] Open
Abstract
The protein p53, known as the "Guardian of the Genome", plays an important role in maintaining DNA integrity, providing protection against cancer-promoting mutations. Dysfunction of p53 is observed in almost every cancer, with 50% of cases bearing loss-of-function mutations/deletions in the TP53 gene. In the remaining 50% of cases the overexpression of HDM2 (mouse double minute 2, human homolog) protein, which is a natural inhibitor of p53, is the most common way of keeping p53 inactive. Disruption of HDM2-p53 interaction with the use of HDM2 antagonists leads to the release of p53 and expression of its target genes, engaged in the induction of cell cycle arrest, DNA repair, senescence, and apoptosis. The induction of apoptosis, however, is restricted to only a handful of p53wt cells, and, generally, cancer cells treated with HDM2 antagonists are not efficiently eliminated. For this reason, HDM2 antagonists were tested in combinations with multiple other therapeutics in a search for synergy that would enhance the cancer eradication. This manuscript aims at reviewing the recent progress in developing strategies of combined cancer treatment with the use of HDM2 antagonists.
Collapse
Affiliation(s)
- Justyna Kocik
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Monika Machula
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Aneta Wisniewska
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Ewa Surmiak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Tad A Holak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Lukasz Skalniak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
52
|
Role of Zinc and Selenium in Oxidative Stress and Immunosenescence: Implications for Healthy Aging and Longevity. HANDBOOK OF IMMUNOSENESCENCE 2019. [PMCID: PMC7121636 DOI: 10.1007/978-3-319-99375-1_66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aging is a complex process that includes gradual and spontaneous biochemical and physiological changes which contributes to a decline in performance and increased susceptibility to diseases. Zn and Se are essential trace elements that play a pivotal role in immune functions and antioxidant defense and, consequently, are claimed to play also a role in successful aging trajectories. Consistently with their nature of essential trace elements, a plethora of data obtained “in vitro” and “in vivo” (in humans and animal models) support the relevance of Zn and Se for both the innate and adoptive immune response. Moreover, Zn and Se are strictly involved in the synthesis and regulation of activity of proteins and enzymes, e.g., metallothioneins (MT) and glutathione peroxidase (GPX), that are necessary for our endogenous antioxidant response. This is clearly important to protect our cells from oxidative damage and to slow the decline of our immune system with aging. Age-related changes affecting tissue levels of Zn and Se may indicate that the risk of Zn and Se deficiency increases with aging. However, it is still unclear which of these changes can be the consequence of a “real deficiency” and which can be part of our physiological compensatory response to the accumulating damage occurring in aging. Furthermore, the upregulation of antioxidant proteins (Zn and Se dependent) may be a manifestation of self-induced oxidative stress. By the way, Zn and Se dependent proteins are modulated not only by nutritional status, but also by well-known hallmarks of aging that play antagonistic functions, such as the deregulated nutrient sensing pathways and cellular senescence. Thus, it is not an easy task to conduct Zn or Se supplementation in elderly and it is emerging consistent that these kind of supplementation requires an individualized approach. Anyway, there is consistent support that supplementation with Zn using doses around 10 mg/day is generally safe in elderly and may even improve part of immune performances in those subjects with a baseline deficiency. Regarding Se supplementation, it may induce both beneficial and detrimental effects on cellular immunity depending on the form of Se, supplemental dose, and delivery matrix. The nutritional association of supplements based on “Zn plus Se” is hypothesized to provide additional benefits, but this will likely need a more complex individualized approach. The improvement of our knowledge around screening and detection of Zn and Se deficiency in aging could lead to substantial benefits in terms of efficacy of nutritional supplements aimed at ameliorate performance and health in aging.
Collapse
|
53
|
Babikir HA, Afjei R, Paulmurugan R, Massoud TF. Restoring guardianship of the genome: Anticancer drug strategies to reverse oncogenic mutant p53 misfolding. Cancer Treat Rev 2018; 71:19-31. [DOI: 10.1016/j.ctrv.2018.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023]
|
54
|
Miller JJ, Orvain C, Jozi S, Clarke RM, Smith JR, Blanchet A, Gaiddon C, Warren JJ, Storr T. Multifunctional Compounds for Activation of the p53-Y220C Mutant in Cancer. Chemistry 2018; 24:17734-17742. [PMID: 30230059 DOI: 10.1002/chem.201802677] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/13/2018] [Indexed: 01/19/2023]
Abstract
The p53 protein plays a major role in cancer prevention, and over 50 % of cancer diagnoses can be attributed to p53 malfunction. The common p53 mutation Y220C causes local protein unfolding, aggregation, and can result in a loss of Zn in the DNA-binding domain. Structural analysis has shown that this mutant creates a surface site that can be stabilized using small molecules, and herein a multifunctional approach to restore function to p53-Y220C is reported. A series of compounds has been designed that contain iodinated phenols aimed for interaction and stabilization of the p53-Y220C surface cavity, and Zn-binding fragments for metallochaperone activity. Their Zn-binding affinity was characterized using spectroscopic methods and demonstrate the ability of compounds L4 and L5 to increase intracellular levels of Zn2+ in a p53-Y220C-mutant cell line. The in vitro cytotoxicity of our compounds was initially screened by the National Cancer Institute (NCI-60), followed by testing in three stomach cancer cell lines with varying p53 status', including AGS (WTp53), MKN1 (V143A), and NUGC3 (Y220C). Our most promising ligand, L5, is nearly 3-fold more cytotoxic than cisplatin in a large number of cell lines. The impressive cytotoxicity of L5 is further maintained in a NUGC3 3D spheroid model. L5 also induces Y220C-specific apoptosis in a cleaved caspase-3 assay, reduces levels of unfolded mutant p53, and recovers p53 transcriptional function in the NUGC3 cell line. These results show that these multifunctional scaffolds have the potential to restore wild-type function in mutant p53-Y220C.
Collapse
Affiliation(s)
- Jessica J Miller
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Christophe Orvain
- Inserm UMR_S 1113, Molecular Mechanisms of Stress Response and Pathologies, Université de Strasbourg, Strasbourg, France
| | - Shireen Jozi
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Ryan M Clarke
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Jason R Smith
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Anaïs Blanchet
- Inserm UMR_S 1113, Molecular Mechanisms of Stress Response and Pathologies, Université de Strasbourg, Strasbourg, France
| | - Christian Gaiddon
- Inserm UMR_S 1113, Molecular Mechanisms of Stress Response and Pathologies, Université de Strasbourg, Strasbourg, France
| | - Jeffrey J Warren
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| |
Collapse
|
55
|
Hacohen N, Ip CJX, Gordon R. Analysis of Egg White Protein Composition with Double Nanohole Optical Tweezers. ACS OMEGA 2018; 3:5266-5272. [PMID: 31458737 PMCID: PMC6641915 DOI: 10.1021/acsomega.8b00651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/03/2018] [Indexed: 05/21/2023]
Abstract
We use a double nanohole optical tweezer to analyze the protein composition of egg white through analysis of many individual protein trapping events. The proteins are grouped by mass based on two metrics: standard deviation of the trapping laser intensity fluctuations from the protein diffusion and the time constant of these fluctuations coming from the autocorrelation. Quantitative analysis is demonstrated for artificial samples, and then, the approach is applied to real egg white. The composition found from real egg white corresponds well to past reports using gel electrophoresis. This approach differs from past works by allowing for individual protein analysis in heterogeneous solutions without the need for denaturing, labeling, or tethering.
Collapse
Affiliation(s)
- Noa Hacohen
- Faculty of Engineering, Department of Electrical and Computer Engineering, University of Victoria, Victoria, British Columbia V8W 3P6, Canada
| | - Candice J X Ip
- Faculty of Engineering, Department of Electrical and Computer Engineering, University of Victoria, Victoria, British Columbia V8W 3P6, Canada
| | - Reuven Gordon
- Faculty of Engineering, Department of Electrical and Computer Engineering, University of Victoria, Victoria, British Columbia V8W 3P6, Canada
| |
Collapse
|
56
|
Kogan S, Carpizo DR. Zinc Metallochaperones as Mutant p53 Reactivators: A New Paradigm in Cancer Therapeutics. Cancers (Basel) 2018; 10:E166. [PMID: 29843463 PMCID: PMC6025018 DOI: 10.3390/cancers10060166] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022] Open
Abstract
Restoration of wild-type structure and function to mutant p53 with a small molecule (hereafter referred to as "reactivating" mutant p53) is one of the holy grails in cancer therapeutics. The majority of TP53 mutations are missense which generate a defective protein that is targetable. We are currently developing a new class of mutant p53 reactivators called zinc metallochaperones (ZMCs) and, here, we review our current understanding of them. The p53 protein requires the binding of a single zinc ion, coordinated by four amino acids in the DNA binding domain, for proper structure and function. Loss of the wild-type structure by impairing zinc binding is a common mechanism of inactivating p53. ZMCs reactivate mutant p53 using a novel two-part mechanism that involves restoring the wild-type structure by reestablishing zinc binding and activating p53 through post-translational modifications induced by cellular reactive oxygen species (ROS). The former causes a wild-type conformation change, the later induces a p53-mediated apoptotic program to kill the cancer cell. ZMCs are small molecule metal ion chelators that bind zinc and other divalent metal ions strong enough to remove zinc from serum albumin, but weak enough to donate it to mutant p53. Recently we have extended our understanding of the mechanism of ZMCs to the role of cells' response to this zinc surge. We found that cellular zinc homeostatic mechanisms, which normally function to maintain free intracellular zinc levels in the picomolar range, are induced by ZMCs. By normalizing zinc levels, they function as an OFF switch to ZMCs because zinc levels are no longer sufficiently high to maintain a wild-type structure. This on/off switch leads to a transient nature to the mechanism of ZMCs in which mutant p53 activity comes on in a few hours and then is turned off. This finding has important implications for the translation of ZMCs to the clinic because it indicates that ZMC concentrations need not be maintained at high levels for their activity. Indeed, we found that short exposures (as little as 15 min) were adequate to observe the mutant p53 reactivating activity. This switch mechanism imparts an advantage over other targeted therapeutics in that efficacy can be accomplished with minimal exposure which minimizes toxicity and maximizes the therapeutic window. This on/off switch mechanism is unique in targeted cancer therapeutics and will impact the design of human clinical trials.
Collapse
Affiliation(s)
- Samuel Kogan
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
- Department of Pharmacology, Rutgers University, Piscataway, NJ 08854, USA.
| | - Darren R Carpizo
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
- Department of Pharmacology, Rutgers University, Piscataway, NJ 08854, USA.
- Z53 Therapeutics, Inc., Holmdel, NJ 07733, USA.
| |
Collapse
|
57
|
Zhao D, Tahaney WM, Mazumdar A, Savage MI, Brown PH. Molecularly targeted therapies for p53-mutant cancers. Cell Mol Life Sci 2017; 74:4171-4187. [PMID: 28643165 PMCID: PMC5664959 DOI: 10.1007/s00018-017-2575-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/30/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
Abstract
The tumor suppressor p53 is lost or mutated in approximately half of human cancers. Mutant p53 not only loses its anti-tumor transcriptional activity, but also often acquires oncogenic functions to promote tumor proliferation, invasion, and drug resistance. Traditional strategies have been taken to directly target p53 mutants through identifying small molecular compounds to deplete mutant p53, or to restore its tumor suppressive function. Accumulating evidence suggest that cancer cells with mutated p53 often exhibit specific functional dependencies on secondary genes or pathways to survive, providing alternative targets to indirectly treat p53-mutant cancers. Targeting these genes or pathways, critical for survival in the presence of p53 mutations, holds great promise for cancer treatment. In addition, mutant p53 often exhibits novel gain-of-functions to promote tumor growth and metastasis. Here, we review and discuss strategies targeting mutant p53, with focus on targeting the mutant p53 protein directly, and on the progress of identifying genes and pathways required in p53-mutant cells.
Collapse
Affiliation(s)
- Dekuang Zhao
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - William M Tahaney
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Michelle I Savage
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
58
|
|
59
|
Lei H, Guo Y, Hu X, Hu C, Hu X, Li H. Reversible Unfolding and Folding of the Metalloprotein Ferredoxin Revealed by Single-Molecule Atomic Force Microscopy. J Am Chem Soc 2017; 139:1538-1544. [DOI: 10.1021/jacs.6b11371] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Hai Lei
- State
Key Laboratory of Precision Measurements Technology and Instruments,
School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, People’s Republic of China
- Department
of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Yabin Guo
- Department
of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Xiaodong Hu
- State
Key Laboratory of Precision Measurements Technology and Instruments,
School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Chunguang Hu
- State
Key Laboratory of Precision Measurements Technology and Instruments,
School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Xiaotang Hu
- State
Key Laboratory of Precision Measurements Technology and Instruments,
School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Hongbin Li
- State
Key Laboratory of Precision Measurements Technology and Instruments,
School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, People’s Republic of China
- Department
of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
60
|
Thayer KM, Quinn TR. p53 R175H hydrophobic patch and H-bond reorganization observed by MD simulation. Biopolymers 2016; 105:176-85. [PMID: 26566695 DOI: 10.1002/bip.22766] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/06/2015] [Accepted: 11/10/2015] [Indexed: 01/28/2023]
Abstract
Molecular dynamics simulations probe the origins of aberrant functionality of R175H p53, which normally prevent tumorigenesis. This hotspot mutation exhibits loss of its essential zinc cofactor, aggregation, and activation of gain of function promoters, characteristics contributing to the loss of normal p53 activity. This study provided molecular level insight into the reorganization of the hydrogen bonding network and the formation of a hydrophobic patch on the surface of the protein. The hydrogen bonding network globally redistributes at the expense of the stability of the β-sandwich structure, and surface residues reorganize to expose a 250 Å(2) hydrophobic patch of residues covering approximately 2% of the solvent accessible surface. These changes could both stabilize the protein in the conformation exposing the patch to solvent to mediate the reported aggregation, and cause a destabilization in the area associated with DNA binding residues to affect the specificity. The development of the patch prior to loss of zinc indicates that stabilizing the patch quickly may prevent zinc loss. Considerations for rational design of small molecule therapeutics in light of the structural insight has been discussed and it suggest the positive ring around the hydrophobic patch and conserved residues may constitute a druggable site.
Collapse
Affiliation(s)
- Kelly M Thayer
- Department of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, NY, 12604.,Department of Chemistry, Hall-Atwater Laboratories, Wesleyan University, Middletown, CT, 06459
| | - Taylor R Quinn
- Department of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, NY, 12604.,Department of Chemistry, University of Notre Dame, Notre Dame, IN, 46556
| |
Collapse
|
61
|
Abstract
TP53 is the most commonly mutated gene in cancer, with over half of all human cancers harboring a mutation in the gene. The p53 protein is a transcription factor that functions as a tumor suppressor, and a subset of its numerous roles include the arrest of proliferation, promotion of DNA repair, and induction of apoptosis in cells with severe DNA damage or stress. The vast majority of p53 mutations are single amino acid substitutions within the DNA binding domain, which either directly impede the protein's ability to bind DNA or destabilize the structure, resulting in misfolding. These missense mutant proteins are found at high levels due to loss of the MDM2 mediated regulation, and consequently serve as potential drug targets. Numerous pharmacological approaches have been investigated to restore wild type p53 function to these mutants (so-called reactivating mutant p53) with some entering in clinical trials while most have failed in early development. Recently, the field of cancer drug development has produced a number of new compounds that continue to advance this field, each with a different mechanism of action. Here we sought to review these compounds and approaches to reactivating mutant p53. Given the large number of patients with missense mutant p53 mutations, reactivating mutant p53 remains a highly sought after goal in developmental therapeutics.
Collapse
Affiliation(s)
- Samuel Kogan
- Rutgers Robert Wood Johnson Medical School, Piscataway Township, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Darren Carpizo
- Rutgers Robert Wood Johnson Medical School, Piscataway Township, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Surgery, Division of Surgical Oncology, Rutgers Robert Wood Johnson Medical School, Piscataway Township, NJ, USA
| |
Collapse
|
62
|
Cordani M, Butera G, Pacchiana R, Donadelli M. Molecular interplay between mutant p53 proteins and autophagy in cancer cells. Biochim Biophys Acta Rev Cancer 2016; 1867:19-28. [PMID: 27871965 DOI: 10.1016/j.bbcan.2016.11.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/14/2016] [Accepted: 11/18/2016] [Indexed: 12/19/2022]
Abstract
An increasing number of studies highlight the role of mutant p53 proteins in cancer cell growth and in the worsening of cancer patients' clinical outcome. Autophagy has been widely recognized as a main biological event involved in both the regulation of cancer cell proliferation and in the response of several anticancer drugs. A thorough analysis of scientific literature underlines the reciprocal interplay between mutant p53 proteins and autophagy regulation. In this review, we analytically summarize recent findings, which indicate that gain-of-function (GOF) mutant p53 proteins counteract the autophagic machinery by various molecular mechanisms including the regulation of AMPK and Akt/mTOR pathways, autophagy-related genes (ATGs), HIF-1α target genes, and the mitochondrial citrate carrier CIC. Moreover, we report that mutant p53 protein stability is affected by lysosome-mediated degradation through macroautophagy or chaperone-mediated autophagy, suggesting the use of autophagy stimulators to counteract mutant p53 oncogenic activity. Finally, we discuss the functional role of the interplay between mutant p53 proteins and autophagy in cancer progression, a fundamental knowledge to design more effective therapies against cancers bearing mutant TP53 gene.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Giovanna Butera
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| |
Collapse
|
63
|
Romualdo GR, Goto RL, Henrique Fernandes AA, Cogliati B, Barbisan LF. Dietary zinc deficiency predisposes mice to the development of preneoplastic lesions in chemically-induced hepatocarcinogenesis. Food Chem Toxicol 2016; 96:280-9. [DOI: 10.1016/j.fct.2016.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/25/2022]
|
64
|
Zinc and zinc-containing biomolecules in childhood brain tumors. J Mol Med (Berl) 2016; 94:1199-1215. [PMID: 27638340 DOI: 10.1007/s00109-016-1454-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
Zinc ions are essential cofactors of a wide range of enzymes, transcription factors, and other regulatory proteins. Moreover, zinc is also involved in cellular signaling and enzymes inhibition. Zinc dysregulation, deficiency, over-supply, and imbalance in zinc ion transporters regulation are connected with various diseases including cancer. A zinc ion pool is maintained by two types of proteins: (i) zinc-binding proteins, which act as a buffer and intracellular donors of zinc and (ii) zinc transporters responsible for zinc fluxes into/from cells and organelles. The decreased serum zinc ion levels have been identified in patients suffering from various cancer diseases, including head and neck tumors and breast, prostate, liver, and lung cancer. On the contrary, increased zinc ion levels have been found in breast cancer and other malignant tissues. Zinc metalloproteomes of a majority of tumors including brain ones are still not yet fully understood. Current knowledge show that zinc ion levels and detection of certain zinc-containing proteins may be utilized for diagnostic and prognostic purposes. In addition, these proteins can also be promising therapeutic targets. The aim of the present work is an overview of the importance of zinc ions, zinc transporters, and zinc-containing proteins in brain tumors, which are, after leukemia, the second most common type of childhood cancer and the second leading cause of death in children after accidents.
Collapse
|
65
|
Bromley D, Bauer MR, Fersht AR, Daggett V. An in silico algorithm for identifying stabilizing pockets in proteins: test case, the Y220C mutant of the p53 tumor suppressor protein. Protein Eng Des Sel 2016; 29:377-90. [PMID: 27503952 PMCID: PMC5001139 DOI: 10.1093/protein/gzw035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 04/23/2016] [Accepted: 04/23/2016] [Indexed: 11/14/2022] Open
Abstract
The p53 tumor suppressor protein performs a critical role in stimulating apoptosis and cell cycle arrest in response to oncogenic stress. The function of p53 can be compromised by mutation, leading to increased risk of cancer; approximately 50% of cancers are associated with mutations in the p53 gene, the majority of which are in the core DNA-binding domain. The Y220C mutation of p53, for example, destabilizes the core domain by 4 kcal/mol, leading to rapid denaturation and aggregation. The associated loss of tumor suppressor functionality is associated with approximately 75 000 new cancer cases every year. Destabilized p53 mutants can be 'rescued' and their function restored; binding of a small molecule into a pocket on the surface of mutant p53 can stabilize its wild-type structure and restore its function. Here, we describe an in silico algorithm for identifying potential rescue pockets, including the algorithm's integration with the Dynameomics molecular dynamics data warehouse and the DIVE visual analytics engine. We discuss the results of the application of the method to the Y220C p53 mutant, entailing finding a putative rescue pocket through MD simulations followed by an in silico search for stabilizing ligands that dock into the putative rescue pocket. The top three compounds from this search were tested experimentally and one of them bound in the pocket, as shown by nuclear magnetic resonance, and weakly stabilized the mutant.
Collapse
Affiliation(s)
- Dennis Bromley
- Division of Biomedical and Health Informatics, Department of Biomedical Informatics and Medical Education, University of Washington, Box SLU-BIME 358047, 850 Republican St, Building C, Seattle, WA 98109-4714, USA
| | - Matthias R Bauer
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Alan R Fersht
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Valerie Daggett
- Division of Biomedical and Health Informatics, Department of Biomedical Informatics and Medical Education, University of Washington, Box SLU-BIME 358047, 850 Republican St, Building C, Seattle, WA 98109-4714, USA Department of Bioengineering, University of Washington, Box 355013, Seattle, WA 98195-5013, USA
| |
Collapse
|
66
|
McCorvie TJ, Kopec J, Pey AL, Fitzpatrick F, Patel D, Chalk R, Shrestha L, Yue WW. Molecular basis of classic galactosemia from the structure of human galactose 1-phosphate uridylyltransferase. Hum Mol Genet 2016; 25:2234-2244. [PMID: 27005423 PMCID: PMC5081055 DOI: 10.1093/hmg/ddw091] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 03/14/2016] [Indexed: 01/22/2023] Open
Abstract
Classic galactosemia is a potentially lethal disease caused by the dysfunction of galactose 1-phosphate uridylyltransferase (GALT). Over 300 disease-associated GALT mutations have been reported, with the majority being missense changes, although a better understanding of their underlying molecular effects has been hindered by the lack of structural information for the human enzyme. Here, we present the 1.9 Å resolution crystal structure of human GALT (hGALT) ternary complex, revealing a homodimer arrangement that contains a covalent uridylylated intermediate and glucose-1-phosphate in the active site, as well as a structural zinc-binding site, per monomer. hGALT reveals significant structural differences from bacterial GALT homologues in metal ligation and dimer interactions, and therefore is a zbetter model for understanding the molecular consequences of disease mutations. Both uridylylation and zinc binding influence the stability and aggregation tendency of hGALT. This has implications for disease-associated variants where p.Gln188Arg, the most commonly detected, increases the rate of aggregation in the absence of zinc likely due to its reduced ability to form the uridylylated intermediate. As such our structure serves as a template in the future design of pharmacological chaperone therapies and opens new concepts about the roles of metal binding and activity in protein misfolding by disease-associated mutants.
Collapse
Affiliation(s)
- Thomas J McCorvie
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Jolanta Kopec
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Angel L Pey
- Department of Physical Chemistry, Faculty of Sciences, University of Granada, Granada E-18071, Spain
| | - Fiona Fitzpatrick
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Dipali Patel
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Rod Chalk
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Leela Shrestha
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Wyatt W Yue
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| |
Collapse
|
67
|
Improved cytotoxicity of pyridyl-substituted thiosemicarbazones against MCF-7 when used as metal ionophores. Biometals 2015; 29:157-70. [DOI: 10.1007/s10534-015-9905-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 12/10/2015] [Indexed: 01/06/2023]
|
68
|
GARUFI ALESSIA, D'ORAZI VALERIO, CRISPINI ALESSANDRA, D'ORAZI GABRIELLA. Zn(II)-curc targets p53 in thyroid cancer cells. Int J Oncol 2015; 47:1241-1248. [PMID: 26314369 PMCID: PMC4583539 DOI: 10.3892/ijo.2015.3125] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/13/2015] [Indexed: 02/06/2023] Open
Abstract
TP53 mutation is a common event in many cancers, including thyroid carcinoma. Defective p53 activity promotes cancer resistance to therapies and a more malignant phenotype, acquiring oncogenic functions. Rescuing the function of mutant p53 (mutp53) protein is an attractive anticancer therapeutic strategy. Zn(II)-curc is a novel small molecule that has been shown to target mutp53 protein in several cancer cells, but its effect in thyroid cancer cells remains unclear. Here, we investigated whether Zn(II)-curc could affect p53 in thyroid cancer cells with both p53 mutation (R273H) and wild-type p53. Zn(II)-curc induced mutp53H273 downregulation and reactivation of wild-type functions, such as binding to canonical target promoters and target gene transactivation. This latter effect was similar to that induced by PRIMA-1. In addition, Zn(II)-curc triggered p53 target gene expression in wild-type p53-carrying cells. In combination treatments, Zn(II)-curc enhanced the antitumor activity of chemotherapeutic drugs, in both mutant and wild-type-carrying cancer cells. Taken together, our data indicate that Zn(II)-curc promotes the reactivation of p53 in thyroid cancer cells, providing in vitro evidence for a potential therapeutic approach in thyroid cancers.
Collapse
Affiliation(s)
- ALESSIA GARUFI
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ‘G. d’Annunzio’, Chieti, Italy
| | - VALERIO D'ORAZI
- Department of Surgical Sciences, Sapienza University, Rome, Italy
| | - ALESSANDRA CRISPINI
- Department of Chemistry and Technologic Chemistry, University of Calabria, Cosenza, Italy
| | - GABRIELLA D'ORAZI
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ‘G. d’Annunzio’, Chieti, Italy
| |
Collapse
|
69
|
Garufi A, Ubertini V, Mancini F, D'Orazi V, Baldari S, Moretti F, Bossi G, D'Orazi G. The beneficial effect of Zinc(II) on low-dose chemotherapeutic sensitivity involves p53 activation in wild-type p53-carrying colorectal cancer cells. J Exp Clin Cancer Res 2015; 34:87. [PMID: 26297485 PMCID: PMC4546314 DOI: 10.1186/s13046-015-0206-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Activation of wild-type p53 in response to genotoxic stress occurs through different mechanisms including protein conformation, posttranslational modifications, and nuclear localization, leading to DNA binding to sequence-specific promoters. Zinc ion plays a crucial role in stabilizing p53/DNA binding to induce canonical target genes. Mutant p53 proteins undergo protein misfolding that can be counteracted by zinc. However, whether zinc supplementation might have a beneficial antitumor effect in wild-type p53-carrying cells in combination with drugs, has not been addressed so far. METHODS In this study we compared the effect of two antitumor treatments: on the one hand wild-type p53-carrying colon cancer cells were treated with low and high doses of chemotherapeutic agent Adriamycin and, on the other hand, Adriamycin was used in combination with ZnCl2. Biochemical and molecular analyses were applied to evaluate p53 activity and biological outcomes in this setting. Finally, the effect of the different combination treatments were applied to assess tumor growth in vivo in tumor xenografts. RESULTS We found that low-dose Adriamycin did not induce p53 activation in wtp53-carrying colon cancer cells, unless in combination with ZnCl2. Mechanistically, ZnCl2 was a key determinant in inducing wtp53/DNA binding and transactivation of target genes in response to low-dose Adriamycin that used alone did not achieve such effects. Finally, in vivo studies, in a model of wtp53 colon cancer xenograft, show that low-dose Adriamycin did not induce tumor regression unless in combination with ZnCl2 that activated endogenous wtp53. CONCLUSIONS These results provide evidence that ZnCl2 might be a valuable adjuvant in chemotherapeutic regimens of colorectal cancer harboring wild-type p53, able to both activate p53 and reduce the amount of drugs for antitumor purposes.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy.
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Valentina Ubertini
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Francesca Mancini
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Roma, Italy.
| | - Valerio D'Orazi
- Department of Surgical Sciences, Sapienza University, 00100, Rome, Italy.
| | - Silvia Baldari
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy.
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Fabiola Moretti
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Roma, Italy.
| | - Gianluca Bossi
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| | - Gabriella D'Orazi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy.
- Translational Research Department, Regina Elena National Cancer Institute, 00158, Rome, Italy.
| |
Collapse
|
70
|
Yu X, Blanden AR, Narayanan S, Jayakumar L, Lubin D, Augeri D, Kimball SD, Loh SN, Carpizo DR. Small molecule restoration of wildtype structure and function of mutant p53 using a novel zinc-metallochaperone based mechanism. Oncotarget 2015; 5:8879-92. [PMID: 25294809 PMCID: PMC4253404 DOI: 10.18632/oncotarget.2432] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
NSC319726 (ZMC1) is a small molecule that reactivates mutant p53 by restoration of WT structure/function to the most common p53 missense mutant (p53-R175H). We investigated the mechanism by which ZMC1 reactivates p53-R175H and provide evidence that ZMC1: 1) restores WT structure by functioning as a zinc-metallochaperone, providing an optimal concentration of zinc to facilitate proper folding; and 2) increases cellular reactive oxygen species that transactivate the newly conformed p53-R175H (via post-translational modifications), inducing an apoptotic program. We not only demonstrate that this zinc metallochaperone function is possessed by other zinc-binding small molecules, but that it can reactivate other p53 mutants with impaired zinc binding. This represents a novel mechanism for an anti-cancer drug and a new pathway to drug mutant p53. Significance: We have elucidated a novel mechanism to restore wild-type structure/function to mutant p53 using small molecules functioning as zinc-metallochaperones. The pharmacologic delivery of a metal ion to restore proper folding of a mutant protein is unique to medicinal chemistry and represents a new pathway to drug mutant p53.
Collapse
Affiliation(s)
- Xin Yu
- Rutgers Cancer Institute of New Jersey, New Jersey. Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. These authors contributed equally to this work
| | - Adam R Blanden
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York. These authors contributed equally to this work
| | - Sumana Narayanan
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Lalithapriya Jayakumar
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - David Lubin
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| | - David Augeri
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Piscataway, New Jersey
| | - S David Kimball
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Piscataway, New Jersey
| | - Stewart N Loh
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| | - Darren R Carpizo
- Rutgers Cancer Institute of New Jersey, New Jersey. Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| |
Collapse
|
71
|
Reactivating mutant p53 using small molecules as zinc metallochaperones: awakening a sleeping giant in cancer. Drug Discov Today 2015. [PMID: 26205328 DOI: 10.1016/j.drudis.2015.07.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tumor protein p53 (TP53) is the most commonly mutated gene in human cancer. The majority of mutations are missense, and generate a defective protein that is druggable. Yet, for decades, the small-molecule restoration of wild-type (WT) p53 function in mutant p53 tumors (so-called p53 mutant 'reactivation') has been elusive to researchers. The p53 protein requires the binding of a single zinc ion for proper folding, and impairing zinc binding is a major mechanism for loss of function in missense mutant p53. Here, we describe recent work defining a new class of drugs termed zinc metallochaperones that restore WT p53 structure and function by restoring Zn(2+) to Zn(2+)-deficient mutant p53.
Collapse
|
72
|
Blanden AR, Yu X, Wolfe AJ, Gilleran JA, Augeri DJ, O'Dell RS, Olson EC, Kimball SD, Emge TJ, Movileanu L, Carpizo DR, Loh SN. Synthetic metallochaperone ZMC1 rescues mutant p53 conformation by transporting zinc into cells as an ionophore. Mol Pharmacol 2015; 87:825-31. [PMID: 25710967 PMCID: PMC4407733 DOI: 10.1124/mol.114.097550] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/20/2015] [Indexed: 11/22/2022] Open
Abstract
p53 is a Zn(2+)-dependent tumor suppressor inactivated in >50% of human cancers. The most common mutation, R175H, inactivates p53 by reducing its affinity for the essential zinc ion, leaving the mutant protein unable to bind the metal in the low [Zn(2+)]free environment of the cell. The exploratory cancer drug zinc metallochaperone-1 (ZMC1) was previously demonstrated to reactivate this and other Zn(2+)-binding mutants by binding Zn(2+) and buffering it to a level such that Zn(2+) can repopulate the defective binding site, but how it accomplishes this in the context of living cells and organisms is unclear. In this study, we demonstrated that ZMC1 increases intracellular [Zn(2+)]free by functioning as a Zn(2+) ionophore, binding Zn(2+) in the extracellular environment, diffusing across the plasma membrane, and releasing it intracellularly. It raises intracellular [Zn(2+)]free in cancer (TOV112D) and noncancer human embryonic kidney cell line 293 to 15.8 and 18.1 nM, respectively, with half-times of 2-3 minutes. These [Zn(2+)]free levels are predicted to result in ∼90% saturation of p53-R175H, thus accounting for its observed reactivation. This mechanism is supported by the X-ray crystal structure of the [Zn(ZMC1)2] complex, which demonstrates structural and chemical features consistent with those of known metal ionophores. These findings provide a physical mechanism linking zinc metallochaperone-1 in both in vitro and in vivo activities and define the remaining critical parameter necessary for developing synthetic metallochaperones for clinical use.
Collapse
Affiliation(s)
- Adam R Blanden
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Xin Yu
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Aaron J Wolfe
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - John A Gilleran
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - David J Augeri
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Ryan S O'Dell
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Eric C Olson
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - S David Kimball
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Thomas J Emge
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Liviu Movileanu
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Darren R Carpizo
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| | - Stewart N Loh
- Department of Biochemistry and Molecular Biology (A.R.B.,S.N.L.) and Department of Neuroscience and Physiology (R.S.O., E.C.O.), State University of New York Upstate Medical University, Syracuse, New York; Rutgers Cancer Institute of New Jersey (X.Y., D.R.C.), Department of Surgery, Rutgers Robert Wood Johnson Medical School (X.Y., D.R.C.), Office of Translational Sciences (J.A.G., D.J.A., S.D.K.), and Department of Chemistry and Chemical Biology (T.J.E.), Rutgers University, New Brunswick, New Jersey; and Department of Physics, Syracuse University, Syracuse, New York (A.J.W., L.M.)
| |
Collapse
|
73
|
Abstract
It has been confirmed through studies using the technique of unbiased sequencing that the TP53 tumour suppressor is the most frequently inactivated gene in cancer. This finding, together with results from earlier studies, provides compelling evidence for the idea that p53 ablation is required for the development and maintenance of tumours. Genetic reconstitution of the function of p53 leads to the suppression of established tumours as shown in mouse models. This strongly supports the notion that p53 reactivation by small molecules could provide an efficient strategy to treat cancer. In this review, we summarize recent advances in the development of small molecules that restore the function of mutant p53 by different mechanisms, including stabilization of its folding by Apr-246, which is currently being tested in a Phase II clinical trial. We discuss several classes of compounds that reactivate wild-type p53, such as Mdm2 inhibitors, which are currently undergoing clinical testing, MdmX inhibitors and molecules targeting factors upstream of Mdm2/X or p53 itself. Finally, we consider the clinical applications of compounds targeting p53 and the p53 pathway.
Collapse
Affiliation(s)
- J Zawacka-Pankau
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - G Selivanova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
74
|
Garufi A, Pucci D, D'Orazi V, Cirone M, Bossi G, Avantaggiati ML, D'Orazi G. Degradation of mutant p53H175 protein by Zn(II) through autophagy. Cell Death Dis 2014; 5:e1271. [PMID: 24874727 PMCID: PMC4044866 DOI: 10.1038/cddis.2014.217] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/12/2014] [Accepted: 04/15/2014] [Indexed: 01/01/2023]
Abstract
TP53, one of the most important oncosuppressors, is frequently mutated in cancer. Several p53 mutant proteins escape proteolytic degradation and are highly expressed in an aberrant conformation often acquiring pro-oncogenic activities that promote tumor progression and resistance to therapy. Therefore, it has been vastly proposed that reactivation of wild-type (wt) function(s) from mutant p53 (mutp53) may have therapeutic significance. We have previously reported that Zn(II) restores a folded conformation from mutp53 misfolding, rescuing wild-type (wt) p53/DNA-binding and transcription activities. However, whether Zn(II) affects mutp53 stability has never been investigated. Here we show that a novel Zn(II) compound induced mutp53 (R175H) protein degradation through autophagy, the proteolytic machinery specifically devoted to clearing misfolded proteins. Accordingly, pharmacological or genetic inhibition of autophagy prevented Zn(II)-mediated mutp53H175 degradation as well as the ability of the Zn(II) compound to restore wtp53 DNA-binding and transcription activity from this mutant. By contrast, inhibition of the proteasome failed to do so, suggesting that autophagy is the main route for p53H175 degradation. Mechanistically, Zn(II) restored the wtp53 ability to induce the expression of the p53 target gene DRAM (damage-regulated autophagy modulator), a key regulator of autophagy, leading to autophagic induction. Accordingly, inhibition of wtp53 transactivation by pifithrin-α (PFT-α) impaired both autophagy and mutp53H175 degradation induced by curcumin-based zinc compound (Zn(II)-curc). Viewed together, our results uncover a novel mechanism employed by Zn(II)-curc to reactivate mutp53H175, which involves, at least in part, induction of mutp53 degradation via wtp53-mediated autophagy.
Collapse
Affiliation(s)
- A Garufi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti, Italy
| | - D Pucci
- Centro di Eccellenza CEMIF.CAL-LASCAMM, CR-INSTM, Department of Chimica e Tecnologie Chimiche, University of Calabria, Cosenza, Italy
| | - V D'Orazi
- Department of Surgical Sciences, Sapienza University, Rome, Italy
| | - M Cirone
- Department of Experimental Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - G Bossi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
| | - M L Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - G D'Orazi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti, Italy
| |
Collapse
|
75
|
Inhibition of aminoglycoside 6'-N-acetyltransferase type Ib by zinc: reversal of amikacin resistance in Acinetobacter baumannii and Escherichia coli by a zinc ionophore. Antimicrob Agents Chemother 2014; 58:4238-41. [PMID: 24820083 DOI: 10.1128/aac.00129-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In vitro activity of the aminoglycoside 6'-N-acetyltransferase type Ib [AAC(6')-Ib] was inhibited by ZnCl2 with a 50% inhibitory concentration (IC50) of 15 μM. Growth of Acinetobacter baumannii or Escherichia coli harboring aac(6')-Ib in cultures containing 8 μg/ml amikacin was significantly inhibited by the addition of 2 μM Zn(2+) in complex with the ionophore pyrithione (ZnPT).
Collapse
|
76
|
Khoo KH, Hoe KK, Verma CS, Lane DP. Drugging the p53 pathway: understanding the route to clinical efficacy. Nat Rev Drug Discov 2014; 13:217-36. [PMID: 24577402 DOI: 10.1038/nrd4236] [Citation(s) in RCA: 568] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumour suppressor p53 is the most frequently mutated gene in human cancer, with more than half of all human tumours carrying mutations in this particular gene. Intense efforts to develop drugs that could activate or restore the p53 pathway have now reached clinical trials. The first clinical results with inhibitors of MDM2, a negative regulator of p53, have shown efficacy but hint at on-target toxicities. Here, we describe the current state of the development of p53 pathway modulators and new pathway targets that have emerged. The challenge of targeting protein-protein interactions and a fragile mutant transcription factor has stimulated many exciting new approaches to drug discovery.
Collapse
Affiliation(s)
| | - Khoo Kian Hoe
- p53 Laboratory (p53Lab), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06, Immunos, 138648 Singapore
| | - Chandra S Verma
- 1] Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix, 138671 Singapore. [2] School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore. [3] Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543 Singapore
| | - David P Lane
- p53 Laboratory (p53Lab), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06, Immunos, 138648 Singapore
| |
Collapse
|
77
|
Gu J, Wang B, Liu Y, Zhong L, Tang Y, Guo H, Jiang T, Wang L, Li Y, Cai L. Murine double minute 2 siRNA and wild-type p53 gene therapy interact positively with zinc on prostate tumours in vitro and in vivo. Eur J Cancer 2014; 50:1184-1194. [PMID: 24447832 DOI: 10.1016/j.ejca.2013.12.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/29/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022]
Abstract
Prostate cancer (PCa) often shows either mutations of the p53 gene or inactivation of the P53 protein. The latter may be due to up-regulation of mouse double minute 2 homologue (MDM2), which functions both as an E3 ubiquitin ligase to degrade P53 protein via the proteasome and an inhibitor of P53 transcriptional activation. Zinc plays a crucial role in stabilizing the P53 DNA binding domain, but PCa cells often lack the ability to accumulate sufficient zinc. In the present study, we explore the optimal approach to retention of P53 function. To restore the defective P53 pathway in PCa, we have explored a combined treatment of Pmp53 [a plasmid containing both mdm2 small interfering RNA (Si-MDM2) and the wild-type p53 gene] with zinc. This treatment retains the wild-type P53 conformation and function in PCa in vitro and in vivo. Combined treatments significantly inhibited tumour xenograft growth, retaining wild-type P53 conformation and enhancing its transcriptional regulation of p21 and bax gene expression, leading to the decreased proliferation and increased apoptosis. These in vivo findings were confirmed by in vitro culture of PCa PC-3 (p53 null) or DU145 (mutant p53) cells and showed a positive effect of the combined therapy on cell cycle arrest and massive apoptosis. Our findings suggest that the combined therapy of Pmp53 with zinc is an effective strategy that may open a new therapeutic avenue in some cancers expressing low levels of zinc and a defective P53 status.
Collapse
Affiliation(s)
- Junlian Gu
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Bo Wang
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, Norman Bethune College of Medicine, Jilin University, Changchun, China; Department of Pathology, Inner Mongolia Forestry General Hospital, Yakeshi, China
| | - Yanan Liu
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Lingzhi Zhong
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yufeng Tang
- Department of Bone and Joint Surgery, First Hospital of Jilin University, Changchun, China
| | - Hua Guo
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Tao Jiang
- Department of Blood and Division of Rheumatology, Jilin Province People's Hospital, Changchun, China
| | - Liwei Wang
- Department of Medical Informatics and Health Administration, School of Public Health, Jilin University, Changchun, China
| | - Yang Li
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, Norman Bethune College of Medicine, Jilin University, Changchun, China.
| | - Lu Cai
- KCHRI at the Department of Pediatrics, The University of Louisville, Louisville, USA; Departments of Radiation Oncology, Pharmacology and Toxicology, The University of Louisville, Louisville, USA.
| |
Collapse
|
78
|
Muller PAJ, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 2014; 25:304-17. [PMID: 24651012 PMCID: PMC3970583 DOI: 10.1016/j.ccr.2014.01.021] [Citation(s) in RCA: 1145] [Impact Index Per Article: 104.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/13/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022]
Abstract
Many different types of cancer show a high incidence of TP53 mutations, leading to the expression of mutant p53 proteins. There is growing evidence that these mutant p53s have both lost wild-type p53 tumor suppressor activity and gained functions that help to contribute to malignant progression. Understanding the functions of mutant p53 will help in the development of new therapeutic approaches that may be useful in a broad range of cancer types.
Collapse
Affiliation(s)
- Patricia A J Muller
- Medical Research Council Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK.
| | - Karen H Vousden
- CR-UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
79
|
Mapping the structural and dynamical features of multiple p53 DNA binding domains: insights into loop 1 intrinsic dynamics. PLoS One 2013; 8:e80221. [PMID: 24324553 PMCID: PMC3855832 DOI: 10.1371/journal.pone.0080221] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 09/30/2013] [Indexed: 11/19/2022] Open
Abstract
The transcription factor p53 regulates cellular integrity in response to stress. p53 is mutated in more than half of cancerous cells, with a majority of the mutations localized to the DNA binding domain (DBD). In order to map the structural and dynamical features of the DBD, we carried out multiple copy molecular dynamics simulations (totaling 0.8 μs). Simulations show the loop 1 to be the most dynamic element among the DNA-contacting loops (loops 1-3). Loop 1 occupies two major conformational states: extended and recessed; the former but not the latter displays correlations in atomic fluctuations with those of loop 2 (~24 Å apart). Since loop 1 binds to the major groove whereas loop 2 binds to the minor groove of DNA, our results begin to provide some insight into the possible mechanism underpinning the cooperative nature of DBD binding to DNA. We propose (1) a novel mechanism underlying the dynamics of loop 1 and the possible tread-milling of p53 on DNA and (2) possible mutations on loop 1 residues to restore the transcriptional activity of an oncogenic mutation at a distant site.
Collapse
|
80
|
Garufi A, Trisciuoglio D, Porru M, Leonetti C, Stoppacciaro A, D’Orazi V, Avantaggiati ML, Crispini A, Pucci D, D’Orazi G. A fluorescent curcumin-based Zn(II)-complex reactivates mutant (R175H and R273H) p53 in cancer cells. J Exp Clin Cancer Res 2013; 32:72. [PMID: 24220325 PMCID: PMC3851540 DOI: 10.1186/1756-9966-32-72] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/04/2013] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Mutations of the p53 oncosuppressor gene are amongst the most frequent aberration seen in human cancer. Some mutant (mt) p53 proteins are prone to loss of Zn(II) ion that is bound to the wild-type (wt) core, promoting protein aggregation and therefore unfolding. Misfolded p53 protein conformation impairs wtp53-DNA binding and transactivation activities, favouring tumor growth and resistance to antitumor therapies. Screening studies, devoted to identify small molecules that reactivate mtp53, represent therefore an attractive anti-cancer therapeutic strategy. Here we tested a novel fluorescent curcumin-based Zn(II)-complex (Zn-curc) to evaluate its effect on mtp53 reactivation in cancer cells. METHODS P53 protein conformation was examined after Zn-curc treatment by immunoprecipitation and immunofluorescence assays, using conformation-specific antibodies. The mtp53 reactivation was evaluated by chromatin-immunoprecipitation (ChIP) and semi-quantitative RT-PCR analyses of wild-type p53 target genes. The intratumoral Zn-curc localization was evaluated by immunofluorescence analysis of glioblastoma tissues of an ortothopic mice model. RESULTS The Zn-curc complex induced conformational change in p53-R175H and -R273H mutant proteins, two of the most common p53 mutations. Zn-curc treatment restored wtp53-DNA binding and transactivation functions and induced apoptotic cell death. In vivo studies showed that the Zn-curc complex reached glioblastoma tissues of an ortothopic mice model, highlighting its ability to crossed the blood-tumor barrier. CONCLUSIONS Our results demonstrate that Zn-curc complex may reactivate specific mtp53 proteins and that may cross the blood-tumor barrier, becoming a promising compound for the development of drugs to halt tumor growth.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | - Daniela Trisciuoglio
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | - Manuela Porru
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | - Carlo Leonetti
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
| | | | - Valerio D’Orazi
- Department of Surgical Sciences, Sapienza University, 00100 Rome, Italy
| | - Maria Laura Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Alessandra Crispini
- Centro di Eccellenza CEMIF.CAL-LASCAMM, CR-INSTM, Department of Chimica e Tecnologie Chimiche, University of Calabria, 87100 Cosenza, Italy
| | - Daniela Pucci
- Centro di Eccellenza CEMIF.CAL-LASCAMM, CR-INSTM, Department of Chimica e Tecnologie Chimiche, University of Calabria, 87100 Cosenza, Italy
| | - Gabriella D’Orazi
- Department of Experimental Oncology, Regina Elena National Cancer Institute, 00159 Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
81
|
Eldar A, Rozenberg H, Diskin-Posner Y, Rohs R, Shakked Z. Structural studies of p53 inactivation by DNA-contact mutations and its rescue by suppressor mutations via alternative protein-DNA interactions. Nucleic Acids Res 2013; 41:8748-59. [PMID: 23863845 PMCID: PMC3794590 DOI: 10.1093/nar/gkt630] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A p53 hot-spot mutation found frequently in human cancer is the replacement of R273 by histidine or cysteine residues resulting in p53 loss of function as a tumor suppressor. These mutants can be reactivated by the incorporation of second-site suppressor mutations. Here, we present high-resolution crystal structures of the p53 core domains of the cancer-related proteins, the rescued proteins and their complexes with DNA. The structures show that inactivation of p53 results from the incapacity of the mutated residues to form stabilizing interactions with the DNA backbone, and that reactivation is achieved through alternative interactions formed by the suppressor mutations. Detailed structural and computational analysis demonstrates that the rescued p53 complexes are not fully restored in terms of DNA structure and its interface with p53. Contrary to our previously studied wild-type (wt) p53-DNA complexes showing non-canonical Hoogsteen A/T base pairs of the DNA helix that lead to local minor-groove narrowing and enhanced electrostatic interactions with p53, the current structures display Watson-Crick base pairs associated with direct or water-mediated hydrogen bonds with p53 at the minor groove. These findings highlight the pivotal role played by R273 residues in supporting the unique geometry of the DNA target and its sequence-specific complex with p53.
Collapse
Affiliation(s)
- Amir Eldar
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel and Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Haim Rozenberg
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel and Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Yael Diskin-Posner
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel and Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Remo Rohs
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel and Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Zippora Shakked
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel and Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA 90089, USA,*To whom correspondence should be addressed. Tel: +972 8 934 2672; Fax: +972 8 934 6278;
| |
Collapse
|
82
|
Tuomela S, Autio R, Buerki-Thurnherr T, Arslan O, Kunzmann A, Andersson-Willman B, Wick P, Mathur S, Scheynius A, Krug HF, Fadeel B, Lahesmaa R. Gene expression profiling of immune-competent human cells exposed to engineered zinc oxide or titanium dioxide nanoparticles. PLoS One 2013; 8:e68415. [PMID: 23894303 PMCID: PMC3718780 DOI: 10.1371/journal.pone.0068415] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/29/2013] [Indexed: 12/31/2022] Open
Abstract
A comprehensive in vitro assessment of two commercial metal oxide nanoparticles, TiO2 and ZnO, was performed using human monocyte-derived macrophages (HMDM), monocyte-derived dendritic cells (MDDC), and Jurkat T cell leukemia-derived cell line. TiO2 nanoparticles were found to be non-toxic whereas ZnO nanoparticles caused dose-dependent cell death. Subsequently, global gene expression profiling was performed to identify transcriptional response underlying the cytotoxicity caused by ZnO nanoparticles. Analysis was done with doses 1 µg/ml and 10 µg/ml after 6 and 24 h of exposure. Interestingly, 2703 genes were significantly differentially expressed in HMDM upon exposure to 10 µg/ml ZnO nanoparticles, while in MDDCs only 12 genes were affected. In Jurkat cells, 980 genes were differentially expressed. It is noteworthy that only the gene expression of metallothioneins was upregulated in all the three cell types and a notable proportion of the genes were regulated in a cell type-specific manner. Gene ontology analysis revealed that the top biological processes disturbed in HMDM and Jurkat cells were regulating cell death and growth. In addition, genes controlling immune system development were affected. Using a panel of modified ZnO nanoparticles, we obtained an additional support that the cellular response to ZnO nanoparticles is largely dependent on particle dissolution and show that the ligand used to modify ZnO nanoparticles modulates Zn2+ leaching. Overall, the study provides an extensive resource of transcriptional markers for mediating ZnO nanoparticle-induced toxicity for further mechanistic studies, and demonstrates the value of assessing nanoparticle responses through a combined transcriptomics and bioinformatics approach.
Collapse
Affiliation(s)
- Soile Tuomela
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- Turku Doctoral Programme of Biomedical Sciences, Turku, Finland
| | - Reija Autio
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Tina Buerki-Thurnherr
- Swiss Federal Laboratories for Material Science and Technology, Laboratory for Materials-Biology Interactions, St. Gallen, Switzerland
| | - Osman Arslan
- Inorganic and Materials Chemistry, University of Cologne, Cologne, Germany
| | - Andrea Kunzmann
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Britta Andersson-Willman
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Peter Wick
- Swiss Federal Laboratories for Material Science and Technology, Laboratory for Materials-Biology Interactions, St. Gallen, Switzerland
| | - Sanjay Mathur
- Inorganic and Materials Chemistry, University of Cologne, Cologne, Germany
| | - Annika Scheynius
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Harald F. Krug
- Swiss Federal Laboratories for Material Science and Technology, Laboratory for Materials-Biology Interactions, St. Gallen, Switzerland
| | - Bengt Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- * E-mail:
| |
Collapse
|
83
|
Identification of interconnected markers for T-cell acute lymphoblastic leukemia. BIOMED RESEARCH INTERNATIONAL 2013; 2013:210253. [PMID: 23956970 PMCID: PMC3727179 DOI: 10.1155/2013/210253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a complex disease, resulting from proliferation of differentially arrested immature T cells. The molecular mechanisms and the genes involved in the generation of T-ALL remain largely undefined. In this study, we propose a set of genes to differentiate individuals with T-ALL from the nonleukemia/healthy ones and genes that are not differential themselves but interconnected with highly differentially expressed ones. We provide new suggestions for pathways involved in the cause of T-ALL and show that network-based classification techniques produce fewer genes with more meaningful and successful results than expression-based approaches. We have identified 19 significant subnetworks, containing 102 genes. The classification/prediction accuracies of subnetworks are considerably high, as high as 98%. Subnetworks contain 6 nondifferentially expressed genes, which could potentially participate in pathogenesis of T-ALL. Although these genes are not differential, they may serve as biomarkers if their loss/gain of function contributes to generation of T-ALL via SNPs. We conclude that transcription factors, zinc-ion-binding proteins, and tyrosine kinases are the important protein families to trigger T-ALL. These potential disease-causing genes in our subnetworks may serve as biomarkers, alternative to the traditional ones used for the diagnosis of T-ALL, and help understand the pathogenesis of the disease.
Collapse
|
84
|
Glucose restriction induces cell death in parental but not in homeodomain-interacting protein kinase 2-depleted RKO colon cancer cells: molecular mechanisms and implications for tumor therapy. Cell Death Dis 2013; 4:e639. [PMID: 23703384 PMCID: PMC3674370 DOI: 10.1038/cddis.2013.163] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor cell tolerance to nutrient deprivation can be an important factor for tumor progression, and may depend on deregulation of both oncogenes and oncosuppressor proteins. Homeodomain-interacting protein kinase 2 (HIPK2) is an oncosuppressor that, following its activation by several cellular stress, induces cancer cell death via p53-dependent or -independent pathways. Here, we used genetically matched human RKO colon cancer cells harboring wt-HIPK2 (HIPK2+/+) or stable HIPK2 siRNA interference (siHIPK2) to investigate in vitro whether HIPK2 influenced cell death in glucose restriction. We found that glucose starvation induced cell death, mainly due to c-Jun NH2-terminal kinase activation, in HIPK2+/+cells compared with siHIPK2 cells that did not die. 1H-nuclear magnetic resonance quantitative metabolic analyses showed a marked glycolytic activation in siHIPK2 cells. However, treatment with glycolysis inhibitor 2-deoxy-𝒟-glucose induced cell death only in HIPK2+/+ cells but not in siHIPK2 cells. Similarly, siGlut-1 interference did not re-establish siHIPK2 cell death under glucose restriction, whereas marked cell death was reached only after zinc supplementation, a condition known to reactivate misfolded p53 and inhibit the pseudohypoxic phenotype in this setting. Further siHIPK2 cell death was reached with zinc in combination with autophagy inhibitor. We propose that the metabolic changes acquired by cells after HIPK2 silencing may contribute to induce resistance to cell death in glucose restriction condition, and therefore be directly relevant for tumor progression. Moreover, elimination of such a tolerance might serve as a new strategy for cancer therapy.
Collapse
|
85
|
Zheng P, Takayama SIJ, Mauk AG, Li H. Single Molecule Force Spectroscopy Reveals That Iron Is Released from the Active Site of Rubredoxin by a Stochastic Mechanism. J Am Chem Soc 2013; 135:7992-8000. [DOI: 10.1021/ja402150q] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Peng Zheng
- Department of Chemistry, University of British Columbia, Vancouver, British
Columbia V6T 1Z1, Canada
| | - Shin-ichi J. Takayama
- Department of Biochemistry and
Molecular Biology and the Center for Blood Research, University of British Columbia, Vancouver, British
Columbia V6T 1Z3, Canada
| | - A. Grant Mauk
- Department of Biochemistry and
Molecular Biology and the Center for Blood Research, University of British Columbia, Vancouver, British
Columbia V6T 1Z3, Canada
| | - Hongbin Li
- Department of Chemistry, University of British Columbia, Vancouver, British
Columbia V6T 1Z1, Canada
| |
Collapse
|
86
|
Merlot AM, Kalinowski DS, Richardson DR. Novel chelators for cancer treatment: where are we now? Antioxid Redox Signal 2013; 18:973-1006. [PMID: 22424293 DOI: 10.1089/ars.2012.4540] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Under normal circumstances, cellular iron levels are tightly regulated due to the potential toxic effects of this metal ion. There is evidence that tumors possess altered iron homeostasis, which is mediated by the perturbed expression of iron-related proteins, for example, transferrin receptor 1, ferritin and ferroportin 1. The de-regulation of iron homeostasis in cancer cells reveals a particular vulnerability to iron-depletion using iron chelators. In this review, we examine the absorption of iron from the gut; its transport, metabolism, and homeostasis in mammals; and the molecular pathways involved. Additionally, evidence for alterations in iron processing in cancer are described along with the perturbations in other biologically important transition metal ions, for example, copper(II) and zinc(II). These changes can be therapeutically manipulated by the use of novel chelators that have recently been shown to be highly effective in terms of inhibiting tumor growth. RECENT ADVANCES Such chelators include those of the thiosemicarbazone class that were originally thought to target only ribonucleotide reductase, but are now known to have multiple effects, including the generation of cytotoxic radicals. CRITICAL ISSUES Several chelators have shown marked anti-tumor activity in vivo against a variety of solid tumors. An important aspect is the toxicology and the efficacy of these agents in clinical trials. FUTURE DIRECTIONS As part of the process of the clinical assessment of the new chelators, an extensive toxicological assessment in multiple animal models is essential for designing appropriate dosing protocols in humans.
Collapse
Affiliation(s)
- Angelica M Merlot
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, Australia
| | | | | |
Collapse
|
87
|
Scotcher J, Clarke DJ, Mackay CL, Hupp T, Sadler PJ, Langridge-Smith PRR. Redox regulation of tumour suppressor protein p53: identification of the sites of hydrogen peroxide oxidation and glutathionylation. Chem Sci 2013. [DOI: 10.1039/c2sc21702c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
88
|
D'Orazi G, Rinaldo C, Soddu S. Updates on HIPK2: a resourceful oncosuppressor for clearing cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:63. [PMID: 22889244 PMCID: PMC3432601 DOI: 10.1186/1756-9966-31-63] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 07/27/2012] [Indexed: 02/04/2023]
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a multitalented protein that exploits its kinase activity to modulate key molecular pathways in cancer to restrain tumor growth and induce response to therapies. HIPK2 phosphorylates oncosuppressor p53 for apoptotic activation. In addition, also p53-independent apoptotic pathways are regulated by HIPK2 and can be exploited for anticancer purpose too. Therefore, HIPK2 activity is considered a central switch in targeting tumor cells toward apoptosis upon genotoxic damage and the preservation and/or restoration of HIPK2 function is crucial for an efficient tumor response to therapies. As a proof of principle, HIPK2 knockdown impairs p53 function, induces chemoresistance, angiogenesis, and tumor growth in vivo, on the contrary, HIPK2 overexpression activates apoptotic pathways, counteracts hypoxia, inhibits angiogenesis, and induces chemosensitivity both in p53-dependent and -independent ways. The role of HIPK2 in restraining tumor development was also confirmed by studies with HIPK2 knockout mice. Recent findings demonstrated that HIPK2 inhibitions do exist in tumors and depend by several mechanisms including HIPK2 cytoplasmic localization, protein degradation, and loss of heterozygosity (LOH), recapitulating the biological outcome obtained by RNA interference studies in tumor cells, such as p53 inactivation, resistance to therapies, apoptosis inhibition, and tumor progression. These findings may lead to new diagnostic and therapeutic approaches for treating cancer patients. This review will focus on the last updates about HIPK2 contribution in tumorigenesis and cancer treatment.
Collapse
Affiliation(s)
- Gabriella D'Orazi
- Department of Medical, Oral, and Biotechnological Sciences, University "G, d'Annunzio", Chieti 66013, Italy.
| | | | | |
Collapse
|
89
|
Abstract
Comment on: Yu X, et al. Cancer Cell 2012; 21:614-25.
Collapse
|
90
|
Naeem A, Fazili NA. Defective protein folding and aggregation as the basis of neurodegenerative diseases: the darker aspect of proteins. Cell Biochem Biophys 2012; 61:237-50. [PMID: 21573992 DOI: 10.1007/s12013-011-9200-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The ability of a polypeptide to fold into a unique, functional, and three-dimensional structure depends on the intrinsic properties of the amino acid sequence, function of the molecular chaperones, proteins, and enzymes. Every polypeptide has a finite tendency to misfold and this forms the darker side of the protein world. Partially folded and misfolded proteins that escape the cellular quality control mechanism have the high tendency to form inter-molecular hydrogen bonding between the same protein molecules resulting in aggregation. This review summarizes the underlying and universal mechanism of protein folding. It also deals with the factors responsible for protein misfolding and aggregation. This article describes some of the consequences of such behavior particularly in the context of neurodegenerative conformational diseases such as Alzheimer's, Parkinson's, Huntington's, amyotrophic lateral sclerosis and other non-neurodegenerative conformational diseases such as cancer and cystic fibrosis etc. This will encourage a more proactive approach to the early diagnosis of conformational diseases and nutritional counseling for patients.
Collapse
Affiliation(s)
- Aabgeena Naeem
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India.
| | | |
Collapse
|
91
|
Calhoun S, Daggett V. Structural effects of the L145Q, V157F, and R282W cancer-associated mutations in the p53 DNA-binding core domain. Biochemistry 2011; 50:5345-53. [PMID: 21561095 DOI: 10.1021/bi200192j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The p53 tumor suppressor is a transcription factor involved in many important signaling pathways, such as apoptosis and cell-cycle arrest. In over half of human cancers, p53 function is compromised by a mutation in its gene. Mutations in the p53 DNA-binding core domain destabilize the structure and reduce DNA-binding activity. We performed molecular dynamics simulations at physiological temperature to study the structural and dynamic effects of the L145Q, V157F, and R282W cancer-associated mutations in comparison to the wild-type protein. While there were common regions of destabilization in the mutant simulations, structural changes particular to individual mutations were also observed. Significant backbone deviations of the H2 helix and S7-S8 loop were observed in all mutant simulations; the H2 helix binds to DNA. In addition, the L145Q and V157F mutations, which are located in the β-sandwich core of the domain, disrupted the β-sheet structure and the loop-sheet-helix motif. The R282W mutation caused distortion of the loop-sheet-helix motif, but otherwise this mutant was similar to the wild-type structure. The introduction of these mutations caused rearrangement of the DNA-binding surface, consistent with their reduced DNA-binding activity. The simulations reveal detailed effects of the mutations on the stability and dynamics of p53 that may provide insight for therapeutic approaches.
Collapse
Affiliation(s)
- Sara Calhoun
- Department of Bioengineering, University of Washington, Seattle, WA 98195-5013, USA
| | | |
Collapse
|
92
|
Berger E, Haller D. Structure-function analysis of the tertiary bile acid TUDCA for the resolution of endoplasmic reticulum stress in intestinal epithelial cells. Biochem Biophys Res Commun 2011; 409:610-5. [PMID: 21605547 DOI: 10.1016/j.bbrc.2011.05.043] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel diseases (IBD) are chronically relapsing and immune-mediated disorders of the gastrointestinal tract. Endoplasmic reticulum (ER) stress mechanisms in the epithelium have been demonstrated to be implemented into the pathogenesis of intestinal inflammation. Chemical chaperones have been demonstrated to exhibit beneficial effects in various diseases associated with ER stress mechanisms by prohibiting the unfolded protein response (UPR). In a structure-function analysis, we tested the potential of the conjugated bile salt sodium tauroursodeoxycholate (TUDCA), naturally present in the small bowel, to resolve ER stress in intestinal epithelial cells. TUDCA efficiently inhibited the expression of UPR dependent genes like GRP78 triggered by the ER stressor tunicamycin in the small intestinal epithelial cell line Mode-K. TUDCA inhibited upstream signaling events in all three branches of the UPR cascade and diminished binding of UPR activated transcription factors to the grp78 promoter. A structure-function analysis revealed that UDCA but not its conjugation partner taurine, known as a chemical chaperone, is responsible for the inhibition of GRP78 induction and that UDCA is 10 times more effective than its taurine conjugate. This inhibitory effect was confirmed in a cell free assay, where TUDCA and UDCA but not taurine effectively inhibited the aggregation of thermally denatured BSA. We conclude that TUDCA and UDCA are potent anti-aggregants for the resolution of ER stress in intestinal epithelial cells and should be considered as a potential drug target to resolve ER stress mechanisms underlying the pathology of IBD.
Collapse
Affiliation(s)
- Emanuel Berger
- Chair for Biofunctionality, ZIEL Research Center for Nutrition and Food Science, CDD Center for Diet and Disease, Technische Universität München, Gregor-Mendel-Str. 2, 85350 Freising-Weihenstephan, Germany
| | | |
Collapse
|
93
|
Paleček E, Ostatná V, Černocká H, Joerger AC, Fersht AR. Electrocatalytic Monitoring of Metal Binding and Mutation-Induced Conformational Changes in p53 at Picomole Level. J Am Chem Soc 2011; 133:7190-6. [DOI: 10.1021/ja201006s] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Emil Paleček
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | - Veronika Ostatná
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | - Hana Černocká
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | | | - Alan R. Fersht
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, U.K
| |
Collapse
|