151
|
Benyettou F, Prakasam T, Ramdas Nair A, Witzel II, Alhashimi M, Skorjanc T, Olsen JC, Sadler KC, Trabolsi A. Potent and selective in vitro and in vivo antiproliferative effects of metal-organic trefoil knots. Chem Sci 2019; 10:5884-5892. [PMID: 31360392 PMCID: PMC6582759 DOI: 10.1039/c9sc01218d] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023] Open
Abstract
A set of metal-organic trefoil knots (M-TKs) generated by metal-templated self-assembly of a simple pair of chelating ligands were well tolerated in vitro by non-cancer cells but were significantly more potent than cisplatin in both human cancer cells--including those resistant to cisplatin--and in zebrafish embryos. In cultured cells, M-TKs generated reactive oxygen species that triggered apoptosis via the mitochondrial pathway without directly disrupting the cell-membrane or damaging nuclear DNA. The cytotoxicity and wide scope for structural variation of M-TKs indicate the potential of synthetic metal-organic knots as a new field of chemical space for pharmaceutical design and development.
Collapse
Affiliation(s)
- Farah Benyettou
- Program in Chemistry , New York University Abu Dhabi , UAE .
| | | | | | | | - Marwa Alhashimi
- Program in Chemistry , New York University Abu Dhabi , UAE .
| | - Tina Skorjanc
- Program in Chemistry , New York University Abu Dhabi , UAE .
| | - John-Carl Olsen
- Department of Chemistry , University of Rochester , Rochester , New York , USA
| | | | - Ali Trabolsi
- Program in Chemistry , New York University Abu Dhabi , UAE .
| |
Collapse
|
152
|
Du E, Hu X, Li G, Zhang S, Mang D, Roy S, Sasaki T, Zhang Y. Self-Assembly-Directed Cancer Cell Membrane Insertion of Synthetic Analogues for Permeability Alteration. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7376-7382. [PMID: 30091933 DOI: 10.1021/acs.langmuir.8b02107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inspired by the metamorphosis of pore-forming toxins from soluble inactive monomers to cytolytic transmembrane assemblies, we developed self-assembly-directed membrane insertion of synthetic analogues for permeability alteration. An expanded π-conjugation-based molecular precursor with an extremely high rigidity and a long hydrophobic length that is comparable to the hydrophobic width of plasma membrane was synthesized for membrane-inserted self-assembly. Guided by the cancer biomarker expression in vitro, the soluble precursors transform into hydrophobic monomers forming assemblies inserted into the fluid phase of the membrane exclusively. Membrane insertion of rigid synthetic analogues destroys the selective permeability of the plasma membrane gradually. It eventually leads to cancer cell death, including drug resistant cancer cells.
Collapse
|
153
|
Zhang C, Yang M, Ericsson AC. Antimicrobial Peptides: Potential Application in Liver Cancer. Front Microbiol 2019; 10:1257. [PMID: 31231341 PMCID: PMC6560174 DOI: 10.3389/fmicb.2019.01257] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 05/21/2019] [Indexed: 01/08/2023] Open
Abstract
The physicochemical properties of antimicrobial peptides (AMPs) including size, net charge, amphipathic structure, hydrophobicity, and mode-of-action together determine their broad-spectrum activities against bacteria, fungi, protozoa, and viruses. Recent studies show that some AMPs have both antimicrobial and anticancer activities, suggesting a new strategy for cancer therapy. Hepatocellular carcinoma (HCC), the primary liver cancer, is a leading cause of cancer mortality worldwide, and lacks effective treatment. Anticancer peptides (ACPs) derived from AMPs or natural resources could be applied to combat HCC directly or as a synergistic treatment. However, the number of known ACPs is low compared to the number of antibacterial and antifungal peptides, and very few of them can be applied clinically for HCC treatment. In this review, we first summarize recent studies related to ACPs for HCC, followed by a description of potential modes-of-action including direct killing, anti-inflammation, immune modulation, and enhanced wound healing. We then describe the structures of AMPs and methods to design and modify these peptides to improve their anticancer efficacy. Finally, we explore the potential application of ACPs as vaccines or nanoparticles for HCC treatment. Overall, ACPs display several attractive properties as therapeutic agents, including broad-spectrum anticancer activity, ease-of-design and modification, and low production costs. As this is an emerging and novel area of cancer therapy, additional studies are needed to identify existing candidate AMPs with ACP activity, and assess their anticancer activity and specificity, and immunomodulatory effects, using in vitro, in silico, and in vivo approaches.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, United States
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
- University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
154
|
Amerikova M, Pencheva El-Tibi I, Maslarska V, Bozhanov S, Tachkov K. Antimicrobial activity, mechanism of action, and methods for stabilisation of defensins as new therapeutic agents. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1611385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Meri Amerikova
- Department of Pharmaceutical Chemistry Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Ivanka Pencheva El-Tibi
- Department of Pharmaceutical Chemistry Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Vania Maslarska
- Department of Chemistry Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Stanislav Bozhanov
- Department of Chemistry Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Konstantin Tachkov
- Department of Social Pharmacy Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
155
|
Yi HC, You ZH, Zhou X, Cheng L, Li X, Jiang TH, Chen ZH. ACP-DL: A Deep Learning Long Short-Term Memory Model to Predict Anticancer Peptides Using High-Efficiency Feature Representation. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:1-9. [PMID: 31173946 PMCID: PMC6554234 DOI: 10.1016/j.omtn.2019.04.025] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 01/10/2023]
Abstract
Cancer is a well-known killer of human beings, which has led to countless deaths and misery. Anticancer peptides open a promising perspective for cancer treatment, and they have various attractive advantages. Conventional wet experiments are expensive and inefficient for finding and identifying novel anticancer peptides. There is an urgent need to develop a novel computational method to predict novel anticancer peptides. In this study, we propose a deep learning long short-term memory (LSTM) neural network model, ACP-DL, to effectively predict novel anticancer peptides. More specifically, to fully exploit peptide sequence information, we developed an efficient feature representation approach by integrating binary profile feature and k-mer sparse matrix of the reduced amino acid alphabet. Then we implemented a deep LSTM model to automatically learn how to identify anticancer peptides and non-anticancer peptides. To our knowledge, this is the first time that the deep LSTM model has been applied to predict anticancer peptides. It was demonstrated by cross-validation experiments that the proposed ACP-DL remarkably outperformed other comparison methods with high accuracy and satisfied specificity on benchmark datasets. In addition, we also contributed two new anticancer peptides benchmark datasets, ACP740 and ACP240, in this work. The source code and datasets are available at https://github.com/haichengyi/ACP-DL.
Collapse
Affiliation(s)
- Hai-Cheng Yi
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhu-Hong You
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China.
| | - Xi Zhou
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Li Cheng
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Xiao Li
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Tong-Hai Jiang
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Zhan-Heng Chen
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| |
Collapse
|
156
|
Biomedical Imaging: Principles, Technologies, Clinical Aspects, Contrast Agents, Limitations and Future Trends in Nanomedicines. Pharm Res 2019; 36:78. [PMID: 30945009 DOI: 10.1007/s11095-019-2608-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022]
Abstract
This review article presents the state-of-the-art in the major imaging modalities supplying relevant information on patient health by real-time monitoring to establish an accurate diagnosis and potential treatment plan. We draw a comprehensive comparison between all imagers and ultimately end with our focus on two main types of scanners: X-ray CT and MRI scanners. Numerous types of imaging probes for both imaging techniques are described, as well as reviewing their strengths and limitations, thereby showing the current need for the development of new diagnostic contrast agents (CAs). The role of nanoparticles in the design of CAs is then extensively detailed, reviewed and discussed. We show how nanoparticulate agents should be promising alternatives to molecular ones and how they are already paving new routes in the field of nanomedicine.
Collapse
|
157
|
Faden F, Mielke S, Dissmeyer N. Modulating Protein Stability to Switch Toxic Protein Function On and Off in Living Cells. PLANT PHYSIOLOGY 2019; 179:929-942. [PMID: 30679267 PMCID: PMC6393803 DOI: 10.1104/pp.18.01215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/10/2019] [Indexed: 05/02/2023]
Abstract
Toxic proteins are prime targets for molecular farming (the generation of pharmacologically active or biotechnologically usable compounds in plants) and are also efficient tools for targeted cell ablation in genetics, developmental biology, and biotechnology. However, achieving conditional activity of cytotoxins and maintaining the toxin-expressing plants as stably transformed lines remain challenging. Here, we produce a switchable version of the highly cytotoxic bacterial RNase barnase by fusing the protein to a portable protein degradation cassette, the low-temperature degron cassette. This method allows conditional genetics based on conditional protein degradation via the N-end rule or N-degron pathway and has been used to vice versa accumulate and/or deplete a diverse variety of highly active, unstable or stable target proteins in different living multicellular organisms and cell systems. Moreover, we expressed the barnase fusion under control of the trichome-specific TRIPTYCHON promoter. This enabled efficient temperature-dependent control of protein accumulation in Arabidopsis (Arabidopsis thaliana) leaf hairs (trichomes). By tuning the levels of the protein, we were able to control the fate of trichomes in vivo. The on-demand formation of trichomes through manipulating the balance between stabilization and destabilization of barnase provides proof of concept for a robust and powerful tool for conditional switchable cell arrest. We present this tool as a potential strategy for the manufacture and accumulation of cytotoxic proteins and toxic high-value products in plants or for conditional genetic cell ablation.
Collapse
Affiliation(s)
- Frederik Faden
- Independent Junior Research Group on Protein Recognition and Degradation, Leibniz Institute of Plant Biochemistry, D-06120 Halle (Saale), Germany
- ScienceCampus Halle, Plant-Based Bioeconomy, D-06120 Halle (Saale), Germany
| | - Stefan Mielke
- Independent Junior Research Group on Protein Recognition and Degradation, Leibniz Institute of Plant Biochemistry, D-06120 Halle (Saale), Germany
- ScienceCampus Halle, Plant-Based Bioeconomy, D-06120 Halle (Saale), Germany
| | - Nico Dissmeyer
- Independent Junior Research Group on Protein Recognition and Degradation, Leibniz Institute of Plant Biochemistry, D-06120 Halle (Saale), Germany
- ScienceCampus Halle, Plant-Based Bioeconomy, D-06120 Halle (Saale), Germany
| |
Collapse
|
158
|
Li Z, Ryszka M, Dawley MM, Carmichael I, Bravaya KB, Ptasińska S. Dipole-Supported Electronic Resonances Mediate Electron-Induced Amide Bond Cleavage. PHYSICAL REVIEW LETTERS 2019; 122:073002. [PMID: 30848645 DOI: 10.1103/physrevlett.122.073002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 10/11/2018] [Indexed: 05/28/2023]
Abstract
Dissociative electron attachment (DEA) plays a key role in radiation damage of biomolecules under high-energy radiation conditions. The initial step in DEA is often rationalized in terms of resonant electron capture into one of the metastable valence states of a molecule followed by its fragmentation. Our combined theoretical and experimental investigations indicate that the manifold of states responsible for electron capture in the DEA process can be dominated by core-excited (shake-up) dipole-supported resonances. Specifically, we present the results of experimental and computational studies of the gas-phase DEA to three prototypical peptide molecules, formamide, N-methylformamide (NMF), and N,N-dimethyl-formamide (DMF). In contrast to the case of electron capture by positively charged peptides in which amide bond rupture is rare compared to N─C_{α} bond cleavage, fragmentation of the amide bond was observed in each of these three molecules. The ion yield curves for ions resulting from this amide bond cleavage, such as NH_{2}^{-} for formamide, NHCH_{3}^{-} for NMF, and N(CH_{3})_{2}^{-} for DMF, showed a double-peak structure in the region between 5 and 8 eV. The peaks are assigned to Feshbach resonances including core-excited dipole-supported resonances populated upon electron attachment based on high-level electronic structure calculations. Moreover, the lower energy peak is attributed to formation of the core-excited resonance that correlates with the triplet state of the neutral molecule. The latter process highlights the role of optically spin-forbidden transitions promoted by electron impact in the DEA process.
Collapse
Affiliation(s)
- Zhou Li
- Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556, USA
- Department of Physics, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Michal Ryszka
- Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - M Michele Dawley
- Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Ian Carmichael
- Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Ksenia B Bravaya
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, USA
| | - Sylwia Ptasińska
- Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556, USA
- Department of Physics, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
159
|
Biswas S, Samui S, Biswas S, Das AK, Naskar J. Molecular recognition of double-stranded DNA by a synthetic, homoaromatic tripeptide (YYY): The spectroscopic and calorimetric study. Int J Biol Macromol 2019; 123:221-227. [DOI: 10.1016/j.ijbiomac.2018.11.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/02/2018] [Accepted: 11/02/2018] [Indexed: 01/02/2023]
|
160
|
Rasoolian M, Kheirollahi M, Hosseini SY. MDA-7/interleukin 24 (IL-24) in tumor gene therapy: application of tumor penetrating/homing peptides for improvement of the effects. Expert Opin Biol Ther 2019; 19:211-223. [PMID: 30612497 DOI: 10.1080/14712598.2019.1566453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION MDA-7/Interleukin-24 (IL-24), as a pleiotropic cytokine, exhibits a specific tumor suppression property that has attracted a great deal of attention. While its anti-tumor induction is mostly attributed to endogenous gene expression, attachment of secreted MDA-7/IL-24 to cognate receptors also triggers the death of cancerous cell via different pathways. Therefore, precise targeting of secreted MDA-7/IL-24 to tumor cells would render it more efficacy and specificity. AREAS COVERED In order to target soluble cytokines, particularly MDA-7/IL-24 to the neighbor tumor sites and enhance their therapeutic efficiency, fusing with cell penetrating peptides (CPPs) or Tumor homing peptides (THPs) seems logical due to the improvement of their bystander effects. Although the detailed anti-tumor mechanisms of endogenous mda-7/IL-24 have been largely investigated, the significance of the secreted form in these activities and methods of its improving by CPPs or THPs need more discussion. EXPERT OPINION While the employment of CPPs/THPs for the improvement of cytokine gene therapy is desirable, to create fusions of CPPs/THPs with MDA-7/IL-24, some hurdles are not avoidable. Regarding our expertise, herein, the importance of CPPs/THPs, needs for their elegant designing in a fusion structure, and their applications in cytokine gene therapy are discussed with a special focus on mda-7/IL-24.
Collapse
Affiliation(s)
- Mohammad Rasoolian
- a Department of Genetics and Molecular Biology, School of Medicine , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Majid Kheirollahi
- a Department of Genetics and Molecular Biology, School of Medicine , Isfahan University of Medical Sciences , Isfahan , Iran.,b Department of Genetics and Molecular Biology, Pediatrics Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease School of Medicine , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Seyed Younes Hosseini
- c Bacteriology and Virology Department, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
161
|
Costa SA, Mozhdehi D, Dzuricky MJ, Isaacs FJ, Brustad EM, Chilkoti A. Active Targeting of Cancer Cells by Nanobody Decorated Polypeptide Micelle with Bio-orthogonally Conjugated Drug. NANO LETTERS 2019; 19:247-254. [PMID: 30540482 PMCID: PMC6465085 DOI: 10.1021/acs.nanolett.8b03837] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Polypeptides are promising carriers for chemotherapeutics: they have minimal toxicity, can be recombinantly synthesized with precise control over molecular weight, and enhance drug pharmacokinetics as self-assembled nanoparticles. Polypeptide-based systems also provide the ability to achieve active targeting with genetically encoded targeting ligands. While passive targeting promotes accumulation of nanocarriers in solid tumors, active targeting provides an additional layer of tunable control and widens the therapeutic window. However, fusion of most targeting proteins to polypeptide carriers exposes the limitations of this approach: the residues that are used for drug attachment are also promiscuously distributed on protein surfaces. We present here a universal methodology to solve this problem by the site-specific attachment of extrinsic moieties to polypeptide drug delivery systems without cross-reactivity to fused targeting domains. We incorporate an unnatural amino acid, p-acetylphenylalanine, to provide a biorthogonal ketone for attachment of doxorubicin in the presence of reactive amino acids in a nanobody-targeted, elastin-like polypeptide nanoparticle. These nanoparticles exhibit significantly greater cytotoxicity than nontargeted controls in multiple cancer cell lines.
Collapse
Affiliation(s)
- Simone A. Costa
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Davoud Mozhdehi
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Michael J. Dzuricky
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Farren J. Isaacs
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06520, United States
| | - Eric M. Brustad
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
162
|
Kostrzewa T, Sahu KK, Gorska-Ponikowska M, Tuszynski JA, Kuban-Jankowska A. Synthesis of small peptide compounds, molecular docking, and inhibitory activity evaluation against phosphatases PTP1B and SHP2. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:4139-4147. [PMID: 30584278 PMCID: PMC6287413 DOI: 10.2147/dddt.s186614] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Background The protein tyrosine phosphatases PTP1B and SHP2 are promising drug targets in treatment design for breast cancer. Searching for specific inhibitors of their activity has recently become the challenge of many studies. Previous work has indicated that the promising PTP inhibitors may be small compounds that are able to bind and interact with amino residues from the binding site. Purpose The main goal of our study was to synthesize and analyze the effect of selected small peptide inhibitors on oncogenic PTP1B and SHP2 enzymatic activity and viability of MCF7 breast cancer cells. We also performed computational analysis of peptides binding with allosteric sites of PTP1B and SHP2 phosphatases. Methods We measured the inhibitory activity of compounds utilizing recombinant enzymes and MCF7 cell line. Computational analysis involved docking studies of binding conformation and interactions of inhibitors with allosteric sites of phosphatases. Results The results showed that the tested compounds decrease the enzymatic activity of phosphatases PTP1B and SHP2 with IC50 values in micromolar ranges. We observed higher inhibitory activity of dipeptides than tripeptides. Phe-Asp was the most effective against SHP2 enzymatic activity, with IC50=5.2±0.4 µM. Micromolar concentrations of tested dipeptides also decreased the viability of MCF7 breast cancer cells, with higher inhibitory activity observed for the Phe-Asp peptide. Moreover, the peptides tested were able to bind and interact with allosteric sites of PTP1B and SHP2 phosphatases. Conclusion Our research showed that small peptide compounds can be considered for the design of specific inhibitors of oncogenic protein tyrosine phosphatases.
Collapse
Affiliation(s)
- Tomasz Kostrzewa
- Department of Medical Chemistry, Medical University of Gdańsk, Gdańsk, Poland,
| | - Kamlesh K Sahu
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | | | - Jack A Tuszynski
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
163
|
Peptide-based targeted therapeutics: Focus on cancer treatment. J Control Release 2018; 292:141-162. [DOI: 10.1016/j.jconrel.2018.11.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 12/14/2022]
|
164
|
Wei L, Zhou C, Chen H, Song J, Su R. ACPred-FL: a sequence-based predictor using effective feature representation to improve the prediction of anti-cancer peptides. Bioinformatics 2018; 34:4007-4016. [PMID: 29868903 PMCID: PMC6247924 DOI: 10.1093/bioinformatics/bty451] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/14/2018] [Accepted: 05/29/2018] [Indexed: 11/15/2022] Open
Abstract
Motivation Anti-cancer peptides (ACPs) have recently emerged as promising therapeutic agents for cancer treatment. Due to the avalanche of protein sequence data in the post-genomic era, there is an urgent need to develop automated computational methods to enable fast and accurate identification of novel ACPs within the vast number of candidate proteins and peptides. Results To address this, we propose a novel predictor named Anti-Cancer peptide Predictor with Feature representation Learning (ACPred-FL) for accurate prediction of ACPs based on sequence information. More specifically, we develop an effective feature representation learning model, with which we can extract and learn a set of informative features from a pool of support vector machine-based models trained using sequence-based feature descriptors. By doing so, the class label information of data samples is fully utilized. To improve the feature representation, we further employ a two-step feature selection technique, resulting in a most informative five-dimensional feature vector for the final peptide representation. Experimental results show that such five features provide the most discriminative power for identifying ACPs than currently available feature descriptors, highlighting the effectiveness of the proposed feature representation learning approach. The developed ACPred-FL method significantly outperforms state-of-the-art methods. Availability and implementation The web-server of ACPred-FL is available at http://server.malab.cn/ACPred-FL. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Leyi Wei
- School of Computer Science and Technology, Tianjin University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Chen Zhou
- School of Computer Science and Technology, Tianjin University, Tianjin, China
| | - Huangrong Chen
- School of Computer Science and Technology, Tianjin University, Tianjin, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology
- Monash Centre for Data Science, Faculty of Information Technology, Monash University, Clayton, VIC 3800, Australia
| | - Ran Su
- School of Computer Software, Tianjin University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
165
|
Natural Products to Fight Cancer: A Focus on Juglans regia. Toxins (Basel) 2018; 10:toxins10110469. [PMID: 30441778 PMCID: PMC6266065 DOI: 10.3390/toxins10110469] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/21/2022] Open
Abstract
Even if cancer represents a burden for human society, an exhaustive cure has not been discovered yet. Low therapeutic index and resistance to pharmacotherapy are two of the major limits of antitumour treatments. Natural products represent an excellent library of bioactive molecules. Thus, tapping into the natural world may prove useful in identifying new therapeutic options with favourable pharmaco-toxicological profiles. Juglans regia, or common walnut, is a very resilient tree that has inhabited our planet for thousands of years. Many studies correlate walnut consumption to beneficial effects towards several chronic diseases, such as cancer, mainly due to the bioactive molecules stored in different parts of the plant. Among others, polyphenols, quinones, proteins, and essential fatty acids contribute to its pharmacologic activity. The present review aims to offer a comprehensive perspective about the antitumour potential of the most promising compounds stored in this plant, such as juglanin, juglone, and the ellagitannin-metabolites urolithins or deriving from walnut dietary intake. All molecules and a chronic intake of the fruit provide tangible anticancer effects. However, the scarcity of studies on humans does not allow results to be conclusive.
Collapse
|
166
|
A Novel Small Peptide Inhibitor of NF κB, RH10, Blocks Oxidative Stress-Dependent Phenotypes in Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5801807. [PMID: 30524659 PMCID: PMC6247396 DOI: 10.1155/2018/5801807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/04/2018] [Indexed: 12/26/2022]
Abstract
Background The RH domain of GRK5 is an effective modulator of cancer growth through the inhibition of NFκB activity. The aim of this study was to identify the minimum effective sequence of RH that is still able to inhibit tumor growth and could be used as a peptide-based drug for therapy. Methods Starting from the RH sequence, small peptides were cloned and tested in KAT-4 cells. The effects on NFκB signaling and its dependent phenotypes were evaluated by Western blot, TUNEL assay, proliferation assay, and angiogenesis in vitro. In vivo experiments were performed in KAT-4 xenografts in Balb/c nude mice. Results A minimum RH ten amino acids long sequence (RH10) was able to interact with IκB, to increase IκB levels, to induce apoptosis, to inhibit KAT4-cell proliferation, NFκB activation, ROS production, and angiogenesis in vitro. In vivo, the peptide inhibited tumor growth in a dose-dependent manner. We also tested its effects in combination with chemotherapeutic drugs and radiotherapy. RH10 ameliorated the antitumor responses to cisplatin, doxorubicin, and ionizing radiation. Conclusion Our data propose RH10 as a potential peptide-based drug to use for cancer treatment both alone or in combination with anticancer therapies.
Collapse
|
167
|
Aptamer Chimeras for Therapeutic Delivery: The Challenging Perspectives. Genes (Basel) 2018; 9:genes9110529. [PMID: 30384431 PMCID: PMC6266988 DOI: 10.3390/genes9110529] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/29/2022] Open
Abstract
Nucleic acid-based aptamers have emerged as efficient delivery carriers of therapeutics. Thanks to their unique features, they can be, to date, considered one of the best targeting moieties, allowing the specific recognition of diseased cells and avoiding unwanted off-target effects on healthy tissues. In this review, we revise the most recent contributes on bispecific and multifunctional aptamer therapeutic chimeras. We will discuss key examples of aptamer-mediated delivery of nucleic acid and peptide-based therapeutics underlying their great potentiality and versatility. Achieved objectives and challenges will be highlighted as well.
Collapse
|
168
|
Smith JD, Cardwell LN, Porciani D, Nguyen JA, Zhang R, Gallazzi F, Tata RR, Burke DH, Daniels MA, Ulery BD. Aptamer-displaying peptide amphiphile micelles as a cell-targeted delivery vehicle of peptide cargoes. Phys Biol 2018; 15:065006. [PMID: 30124431 DOI: 10.1088/1478-3975/aadb68] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Peptide amphiphile micelles (PAMs) are attractive vehicles for the delivery of a variety of therapeutic and prophylactic peptides. However, a key limitation of PAMs is their lack of preferential targeting ability. In this paper, we describe our design of a PAM system that incorporates a DNA oligonucleotide amphiphile (antitail amphiphile-AA) to form A/PAMs. A cell-targeting DNA aptamer with a 3' extension sequence (tail) complementary to the AA is annealed to the surface to form aptamer-displaying PAMs (Aptamer~A/PAMs). Aptamer~A/PAMs are small, anionic, stable nanoparticles capable of delivering a large mass percentage peptide amphiphile (PA) compared to targeting DNA components. Aptamer~A/PAMs are stable for over 4 h in the presence of biological fluids. Additionally, the aptamer retains its cell-targeting properties when annealed to the A/PAM, thus leading to enhanced delivery to a specifically-targeted B-cell leukemia cell line. This exciting modular technology can be readily used with a library of different targeting aptamers and PAs, capable of improving the bioavailability and potency of the peptide cargo.
Collapse
Affiliation(s)
- Josiah D Smith
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Kamal H, Jafar S, Mudgil P, Murali C, Amin A, Maqsood S. Inhibitory properties of camel whey protein hydrolysates toward liver cancer cells, dipeptidyl peptidase-IV, and inflammation. J Dairy Sci 2018; 101:8711-8720. [DOI: 10.3168/jds.2018-14586] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/17/2018] [Indexed: 01/12/2023]
|
170
|
Maraming P, Klaynongsruang S, Boonsiri P, Maijaroen S, Daduang S, Chung JG, Daduang J. Antitumor activity of RT2 peptide derived from crocodile leukocyte peptide on human colon cancer xenografts in nude mice. ENVIRONMENTAL TOXICOLOGY 2018; 33:972-977. [PMID: 30019842 DOI: 10.1002/tox.22584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
RT2, derived from the leukocyte peptide of Crocodylus siamensis, can kill human cervical cancer cells via apoptosis induction, but no evidence has shown in vivo. In this study, we investigated the antitumor effect of RT2 on human colon cancer xenografts in nude mice. Twenty-four mice were injected subcutaneously with human colon cancer HCT 116 cells. Eleven days after cancer cell implantation, the mice were treated with intratumoral injections of phosphate buffered saline (PBS) or RT2 (0.01, 0.1, and 1 mg/mouse) once every 2 days for a total of 5 times. The effect of a 10-day intratumoral injection of RT2 on body weight, biochemical, and hematological parameters in BALB/c mice showed no significant difference between the groups. Tumor volume showed a significant decrease only in the treatment group with RT2 (1 mg/mouse) at day 6 (P < .05), day 8 (P < .01), and day 10 (P < .01) after the first treatment. The protein expression levels of cleaved poly (ADP-ribose) polymerase (PARP), apoptosis-inducing factor (AIF), and the p53 tumor suppressor protein (p53) in xenograft tumors increased after treatment with RT2 (1 mg/mouse) compared to those in the PBS-injected group. Moreover, RT2 increased the expression of Endo G and Bcl-2 family proteins. Therefore, the peptide RT2 can inhibit tumor growth via the induction of apoptosis in an in vivo xenograft model.
Collapse
Affiliation(s)
- Pornsuda Maraming
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Sompong Klaynongsruang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Patcharee Boonsiri
- Faculty of Medicine, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Surachai Maijaroen
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Sakda Daduang
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Science, Khon Kaen University, Khon Kaen, Thailand
| | - Jing-Gung Chung
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Jureerut Daduang
- Center for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
171
|
Maijaroen S, Jangpromma N, Daduang J, Klaynongsruang S. KT2 and RT2 modified antimicrobial peptides derived from Crocodylus siamensis Leucrocin I show activity against human colon cancer HCT-116 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 62:164-176. [PMID: 30031283 DOI: 10.1016/j.etap.2018.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 06/08/2023]
Abstract
Conventional colon cancer treatments have been associated with side effects. Consequently, the discovery of novel effective and safe therapies is urgently needed. Hence, cationic antimicrobial peptides KT2 and RT2 were evaluated towards human colon cancer HCT-116 cells. The MTT assay indicated that both KT2 and RT2 exhibited anticancer activity with good therapeutic indices, and were found to be non-toxic to non-cancerous Vero cells. The IC50 values of KT2 were determined as 111.96 and 90.25 μg/mL while RT2 showed IC50 as 104.07 and 87.84 μg/mL after 12 and 24 h treatments, respectively. Moreover, KT2 and RT2 treatment caused a significant reduction in PI3K, AKT1 and mTOR mRNA expression levels, which resulted in suppression either of HCT-116 proliferation or migration. The mechanism involved in apoptosis induction were due to decreased Bcl-2 and XIAP and increased p53, cytochrome c, caspase-2, caspase-3, caspase-8, and caspase-9 mRNA expression levels. These effects increased the level of cell cycle associated gene p21 and decreased cyclin B1 and cyclin D1 expression.
Collapse
Affiliation(s)
- Surachai Maijaroen
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nisachon Jangpromma
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; Department of Integrated Science, Forensic Science Program, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Jureerut Daduang
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sompong Klaynongsruang
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand; Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
172
|
Huo Y, Xv R, Ma H, Zhou J, Xi X, Wu Q, Duan J, Zhou M, Chen T. Identification of <10 KD peptides in the water extraction of Venenum Bufonis from Bufo gargarizans using Nano LC–MS/MS and De novo sequencing. J Pharm Biomed Anal 2018; 157:156-164. [DOI: 10.1016/j.jpba.2018.05.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/19/2022]
|
173
|
Rehman NU, Abed RMM, Hussain H, Khan HY, Khan A, Khan AL, Ali M, Al-Nasri A, Al-Harrasi K, Al-Rawahi AN, Wadood A, Al-Rawahi A, Al-Harrasi A. Anti-proliferative potential of cyclotetrapeptides from Bacillus velezensis RA5401 and their molecular docking on G-Protein-Coupled Receptors. Microb Pathog 2018; 123:419-425. [PMID: 30075241 DOI: 10.1016/j.micpath.2018.07.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/29/2018] [Accepted: 07/30/2018] [Indexed: 10/28/2022]
Abstract
Elucidation of bioactive chemical compounds from rhizobacteria is highly utilized in pharmaceuticals and naturopathy, due to their health benefits to human and plants. In current study, four cyclopeptides along with one phenyl amide were isolated from the ethyl acetate extract of Bacillus velezensis sp. RA5401. Their structures were determined and characterized as cycle (L-prolyl-L-leucyl)2 (1), cyclo (L-prolyl-l-valine)2 (2), cycle (L-phenylanalyl-L-propyl)2 (3), cyclo (D-pro-L-tyr-L-pro-L-tyr)2 (4) and N-(2-phenylethyl)acetamide (5) on the basis of electron spray ionization mass spectrometry (ESI-MS), nuclear magnetic resonance (NMR) techniques and comparison with the literature data. The five compounds have been isolated for the first time from this species. The effect of various concentrations of these compounds on the proliferation of MDA-MB-231 breast cancer cells was examined. It was found that 1 and 2 induced concentration-independent anti-proliferative effects, while 3, 4 and 5 inhibited cancer cell proliferation in a concentration-dependent manner. Furthermore, to determine the suitable binding targets of these compounds within cancer cell line, detailed target prediction and comparative molecular-docking studies were performed. The compounds 1 and 2 hit intracellular anti-cancer targets of proteases family, while compounds 3, 4 and 5 interacted with different membrane receptors of G-Protein-Coupled Receptors (GPCRs). In conclusion, the Bacillus velezensis RA5401 can be an ideal strain to produce anti-proliferative constituents at industrial scale.
Collapse
Affiliation(s)
- Najeeb Ur Rehman
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Raeid M M Abed
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Hidayat Hussain
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Husain Yar Khan
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Abdul L Khan
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Majid Ali
- Department of Chemistry, COMSATS Institute of Information Technology, Abbottabad, 22060, Pakistan
| | - Abdullah Al-Nasri
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Khalid Al-Harrasi
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Ahmed N Al-Rawahi
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Ahmed Al-Rawahi
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa, 616, Oman.
| |
Collapse
|
174
|
Esmati N, Maddirala AR, Hussein N, Amawi H, Tiwari AK, Andreana PR. Efficient syntheses and anti-cancer activity of xenortides A-D including ent/epi-stereoisomers. Org Biomol Chem 2018; 16:5332-5342. [PMID: 29999086 DOI: 10.1039/c8ob00452h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A one-pot, two-step, total synthesis of naturally occurring xenortides A, B, C and D, (Xens A-D) isolated from the bacterium Xenorhabdus nematophila, and an entire complementary set of stereoisomers, has been achieved. Compounds were synthesized utilizing an isocyanide-based Ugi 4-CR followed by facile N-Boc deprotection. The reaction sequence took advantage of the chiral pool of N-Boc protected amino acids (l-Leu/Val and d-Leu/Val) with aryl isocyanides, phenyl acetaldehyde and methylamine giving the desired Xens A-D (A and B >98% ee) and all subsequent stereoisomers in reasonable yields upon deprotection followed by separation of diastereomers. Also, detailed mechanistic insights for diastereoselectivity of (-)-Xen A, as a model in the Ugi 4-CR, has been described. Moreover, for the first time, this focused library was screened for cytotoxicity against a panel of epithelial cancer cell lines as well as normal cell lines with an MTT proliferation assay. The structure-activity relationship (SAR) study demonstrated that tryptamides Xen B and D were more active than phenylethylamides Xen A and C. Furthermore, (-)-Xen B (IC50 = 19-25 μM) and ent-(+)-Xen D (IC50 = 21-26 μM) gave the highest cytotoxicity and they were also found to be non-toxic toward normal cells. Importantly, the SAR results indicate that the stereochemistry at C8 and C11 in (-)-Xen B and ent-(+)-Xen D play a critical role in cytotoxic activity.
Collapse
Affiliation(s)
- N Esmati
- Department of Chemistry and Biochemistry and School of Green Chemistry and Engineering, The University of Toledo, 2801 W. Bancroft St, Toledo, OH 43606, USA.
| | | | | | | | | | | |
Collapse
|
175
|
The Biological and Biophysical Properties of the Spider Peptide Gomesin. Molecules 2018; 23:molecules23071733. [PMID: 30012962 PMCID: PMC6099743 DOI: 10.3390/molecules23071733] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/11/2018] [Accepted: 07/12/2018] [Indexed: 01/31/2023] Open
Abstract
This review summarises the current knowledge of Gomesin (Gm), an 18-residue long, cationic anti-microbial peptide originally isolated from the haemocytes of the Brazilian tarantula Acanthoscurria gomesiana. The peptide shows potent cytotoxic activity against clinically relevant microbes including Gram-positive and Gram-negative bacteria, fungi, and parasites. In addition, Gm shows in-vitro and in-vivo anti-cancer activities against several human and murine cancers. The peptide exerts its cytotoxic activity by permeabilising cell membranes, but the underlying molecular mechanism of action is still unclear. Due to its potential as a therapeutic agent, the structure and membrane-binding properties, as well as the leakage and cytotoxic activities of Gm have been studied using a range of techniques. This review provides a summary of these studies, with a particular focus on biophysical characterisation studies of peptide variants that have attempted to establish a structure-activity relationship. Future studies are still needed to rationalise the binding affinity and cell-type-specific selectivity of Gm and its variants, while more pre-clinical studies are required to develop Gm into a therapeutically useful peptide.
Collapse
|
176
|
Li B, Lyu P, Xi X, Ge L, Mahadevappa R, Shaw C, Kwok HF. Triggering of cancer cell cycle arrest by a novel scorpion venom-derived peptide-Gonearrestide. J Cell Mol Med 2018; 22:4460-4473. [PMID: 29993185 PMCID: PMC6111814 DOI: 10.1111/jcmm.13745] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
In this study, a novel scorpion venom-derived peptide named Gonearrestide was identified in an in-house constructed scorpion venom library through a combination of high-throughput NGS transcriptome and MS/MS proteome platform. In total, 238 novel peptides were discovered from two scorpion species; and 22 peptides were selected for further study after a battery of functional prediction analysis. Following a series of bioinformatics analysis alongside with in vitro biological functional screenings, Gonearrestide was found to be a highly potent anticancer peptide which acts on a broad spectrum of human cancer cells while causing few if any observed cytotoxic effects on epithelial cells and erythrocytes. We further investigated the precise anticancer mechanism of Gonearrestide by focusing on its effects on the colorectal cancer cell line, HCT116. NGS RNA sequencing was employed to obtain full gene expression profiles in HCT116 cells, cultured in the presence and absence of Gonearrestide, to dissect signalling pathway differences. Taken together the in vitro, in vivo and ex vivo validation studies, it was proven that Gonearrestide could inhibit the growth of primary colon cancer cells and solid tumours by triggering cell cycle arrest in G1 phase through inhibition of cyclin-dependent kinases 4 (CDK4) and up-regulate the expression of cell cycle regulators/inhibitors-cyclin D3, p27, and p21. Furthermore, prediction of signalling pathways and potential binding sites used by Gonearrestide are also presented in this study.
Collapse
Affiliation(s)
- Bin Li
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao
| | - Peng Lyu
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao
| | - Xinping Xi
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao.,School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Lilin Ge
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao.,School of Pharmacy, Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Qixia District, Nanjing, China
| | | | - Chris Shaw
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao
| |
Collapse
|
177
|
Mozayan A, Khaled A. Elucidation of Therapeutic Peptide Binding Partners from Isolated Mitochondria. Cureus 2018; 10:e2898. [PMID: 30397556 PMCID: PMC6207275 DOI: 10.7759/cureus.2898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
CT20p is a protein derived from the C-terminus of Bax. It has selective cytotoxicity for cancer cells, such as the sensitive triple-negative MDA-MB-231 breast adenocarcinoma cells, but not normal cells like the resistant MCF-10A epithelial breast cells. To understand the reason for the peptide’s selective toxicity, a "pull-down" experiment with biotinylated CT20p (biotin-CT20p) and whole-cell protein lysates from breast cancer and normal cells were performed. These studies revealed that CT20p binds to a cytosolic protein called chaperonin-containing TCP-1 (CCT), a molecular chaperone that folds actin and tubulin. However, this method could not detect possible rare interactions made by CT20p with mitochondrial proteins. To determine whether CT20p is associated with mitochondrial proteins as part of the mechanism by which it induces cell death, mitochondrial protein lysates from MDA-MB-231 and MCF-10A cells were isolated and a streptavidin-agarose pulldown procedure using biotin-CT20p was performed. Protein interactions were visualized by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) using silver staining. The results of the experimental procedure showed that biotin-CT20p did not "pull down" any observable mitochondrial proteins from the sensitive MDA-MB-231 cells, indicating that the peptide may not interact with mitochondrial proteins in breast cancer cells. Rather, the interactions observed with biotin-CT20p were with mitochondrial proteins derived from resistant MCF-10A cells, indicating that these interactions were not driving the cancer-selective cell death process. The absence of CT20p-associated proteins from the mitochondrial lysates of MDA-MB-231 breast cancer cells supports the hypothesis that CT20p, unlike the parent protein, Bax, exerts its cytotoxic effects via a cytosolic protein.
Collapse
Affiliation(s)
- Ali Mozayan
- College of Medicine, University of Central Florida, Orlando, USA
| | - Annette Khaled
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, USA
| |
Collapse
|
178
|
Mohammadi-Farsani A, Habibi-Roudkenar M, Golkar M, Shokrgozar MA, Jahanian-Najafabadi A, KhanAhmad H, Valiyari S, Bouzari S. A-NGR fusion protein induces apoptosis in human cancer cells. EXCLI JOURNAL 2018; 17:590-597. [PMID: 30108463 PMCID: PMC6088213 DOI: 10.17179/excli2018-1120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/04/2018] [Indexed: 12/28/2022]
Abstract
The NGR peptide is one of the well-known peptides for targeting tumor cells. It has the ability to target aminopeptidase N (CD13) on tumor cells or the tumor vascular endothelium. In this study, the NGR peptide was used for targeting A subunit of the Shiga toxin to cancer cells. The cytotoxic effect of the A-NGR fusion protein was assessed on HT1080, U937, HT29 cancer cells and MRC-5 normal cells. For this purpose, cells were treated with different concentrations of A-NGR (0.5-40 µg/ml). The evaluation of cell viability was achieved by MTT assay. Apoptosis was determined by annexin-V/PI double staining flow cytometry. Alterations in the mRNA expression of apoptosis - related genes were assessed by real time RT- PCR. The results showed that A-NGR fusion protein effectively inhibited the growth of HT1080 and U937 cancer cells in comparison to negative control (PBS) but for CD13-negative HT-29 cancer cells, only at high concentrations of fusion protein was inhibited growth recorded. On the other hand, A-NGR had little cytotoxic effect on MRC-5 normal cells. The flow cytometry results showed that A-NGR induces apoptosis. Furthermore, the results of real time RT-PCR revealed that A-NGR significantly increases the mRNA expression of caspase 3 and caspase 9. Conclusively, A-NGR fusion protein has the ability of targeting CD13-positive cancer cells, the cytotoxic effect on CD13-positive cancer cells as well as has low cytotoxic effect on normal cells.
Collapse
Affiliation(s)
| | - Mehryar Habibi-Roudkenar
- Medical Biotechnology Department, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Majid Golkar
- Molecular Parasitology Laboratory, Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein KhanAhmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samira Valiyari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
179
|
Peptide chemistry encounters nanomedicine: recent applications and upcoming scenarios in cancer. Future Med Chem 2018; 10:1877-1880. [PMID: 29921135 DOI: 10.4155/fmc-2018-0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
180
|
Novel NGR anchored pullulan micelles for controlled and targeted delivery of doxorubicin to HeLa cancerous cells. IRANIAN POLYMER JOURNAL 2018. [DOI: 10.1007/s13726-018-0606-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
181
|
MUC1 aptamer-targeted DNA micelles for dual tumor therapy using doxorubicin and KLA peptide. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:685-697. [PMID: 29317345 DOI: 10.1016/j.nano.2017.12.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/03/2017] [Accepted: 12/18/2017] [Indexed: 01/04/2023]
Abstract
Targeted delivery of DNA nanoparticles is a promising approach in cancer therapy. Using aptamers, target specific delivery of DNA nanoparticles can be achieved. Further, aptamers can indirectly improve drug encapsulation efficiency of DNA nanoparticles for drugs intercalated within nucleic acid base pairs. Using DNA blocks, a micellar hybrid nanoparticle was prepared for the targeted co-delivery of doxorubicin and a pro-apoptotic peptide, KLA to tumor cells. Results demonstrated that anti-MUC1 aptamer could specifically deliver the synthesized DNA micelle into MCF-7 cells by improving its cellular uptake. Additionally, co-delivery of doxorubicin and KLA could significantly enhance the therapeutic efficacy of the construct resulting in reduction of required dose of doxorubicin that is a pivotal point in reducing chemotherapeutics side effects. Moreover, DOX-KLA-anti-MUC1-micelle remarkably inhibited tumor growth of tumor-bearing mice when compared with free drug. DOX-KLA-anti-MUC1-micelle also reduced toxic effect of free doxorubicin as determined by percent of body weight loss and survival rate in vivo.
Collapse
|
182
|
Ghosh D, Peng X, Leal J, Mohanty R. Peptides as drug delivery vehicles across biological barriers. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018; 48:89-111. [PMID: 29963321 PMCID: PMC6023411 DOI: 10.1007/s40005-017-0374-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/25/2017] [Indexed: 12/15/2022]
Abstract
Peptides are small biological molecules that are attractive in drug delivery and materials engineering for applications including therapeutics, molecular building blocks and cell-targeting ligands. Peptides are small but can possess complexity and functionality as larger proteins. Due to their intrinsic properties, peptides are able to overcome the physiological and transport barriers presented by diseases. In this review, we discuss the progress of identifying and using peptides to shuttle across biological barriers and facilitate transport of drugs and drug delivery systems for improved therapy. Here, the focus of this review is on rationally designed, phage display peptides, and even endogenous peptides as carriers to penetrate biological barriers, specifically the blood-brain barrier(BBB), the gastrointestinal tract (GI), and the solid tumor microenvironment (T). We will discuss recent advances of peptides as drug carriers in these biological environments. From these findings, challenges and potential opportunities to iterate and improve peptide-based approaches will be discussed to translate their promise towards the clinic to deliver drugs for therapeutic efficacy.
Collapse
Affiliation(s)
- Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| | - Xiujuan Peng
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| | - Jasmim Leal
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| | - Rashmi Mohanty
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| |
Collapse
|
183
|
Niu W, Teng IT, Chen X, Tan W, Veige AS. Aptamer-mediated selective delivery of a cytotoxic cationic NHC-Au(i) complex to cancer cells. Dalton Trans 2017; 47:120-126. [PMID: 29192701 PMCID: PMC5736135 DOI: 10.1039/c7dt02616a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A novel cationic NHC-Au(i) complex was synthesized and studied for its antitumor activity. For all the cell lines tested, cationic NHC-Au(i) complex 2 shows much higher cytotoxicity than its neutral analogue 1. To achieve selective cancer cell targeting, complex 2 was covalently conjugated to aptamer AS1411, a DNA aptamer with strong binding affinity for nucleolin. The successful conjugation was confirmed by HPLC, gel electrophoresis, fluorescence spectroscopy and UV-Vis absorption. Conjugate AS1411-2 was then examined for its specific targeting and binding ability towards cancer cells over human normal cells using flow cytometry analysis and confocal microscopy. The cytotoxicity of AS1411-2 was then estimated by MTS assay. It was found that AS1411-2 exhibits higher activity than complex 2 towards targeted cells. Importantly, AS1411-2 exhibits much lower cytotoxicity towards healthy normal cell lines. Concurrently, the control groups without the AS1411 aptamer or without the NHC-Au(i) complex do have significant impact on cancer cell viability.
Collapse
Affiliation(s)
- Weijia Niu
- University of Florida, Department of Chemistry, Center for Catalysis, P.O. Box 117200, Gainesville, Florida 32611, USA.
| | | | | | | | | |
Collapse
|
184
|
Liu X, Jiang J, Ji Y, Lu J, Chan R, Meng H. Targeted drug delivery using iRGD peptide for solid cancer treatment. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2017; 2:370-379. [PMID: 30498580 PMCID: PMC6258069 DOI: 10.1039/c7me00050b] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Many solid tumor types, such as pancreatic cancer, have a generally poor prognosis, in part because the delivery of therapeutic regimen is prohibited by pathological abnormalities that block access to tumor vasculature, leading to poor bioavailability. Recent development of tumor penetrating iRGD peptide that is covalently conjugated on nanocarriers' surface or co-administered with nanocarriers becomes a popular approach for tumor targeting. More importantly, scientists have unlocked an important tumor transcytosis mechanism by which drug carrying nanoparticles directly access solid tumors (without the need of leaky vasculature), thereby allowing systemically injected nanocarriers more abundantly distribute at tumor site with improved efficacy. In this focused review, we summarized the design and implementation strategy for iRGD-mediated tumor targeting. This includes the working principle of such peptide and discussion on patient-specific iRGD effect in vivo, commensurate with the level of key biomarker (i.e. neuropilin-1) expression on tumor vasculature. This highlights the necessity to contemplate the use of a personalized approach when iRGD technology is used in clinic.
Collapse
Affiliation(s)
- Xiangsheng Liu
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Ying Ji
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Jianqin Lu
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Ryan Chan
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Huan Meng
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
185
|
Manavalan B, Basith S, Shin TH, Choi S, Kim MO, Lee G. MLACP: machine-learning-based prediction of anticancer peptides. Oncotarget 2017; 8:77121-77136. [PMID: 29100375 PMCID: PMC5652333 DOI: 10.18632/oncotarget.20365] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/13/2017] [Indexed: 01/25/2023] Open
Abstract
Cancer is the second leading cause of death globally, and use of therapeutic peptides to target and kill cancer cells has received considerable attention in recent years. Identification of anticancer peptides (ACPs) through wet-lab experimentation is expensive and often time consuming; therefore, development of an efficient computational method is essential to identify potential ACP candidates prior to in vitro experimentation. In this study, we developed support vector machine- and random forest-based machine-learning methods for the prediction of ACPs using the features calculated from the amino acid sequence, including amino acid composition, dipeptide composition, atomic composition, and physicochemical properties. We trained our methods using the Tyagi-B dataset and determined the machine parameters by 10-fold cross-validation. Furthermore, we evaluated the performance of our methods on two benchmarking datasets, with our results showing that the random forest-based method outperformed the existing methods with an average accuracy and Matthews correlation coefficient value of 88.7% and 0.78, respectively. To assist the scientific community, we also developed a publicly accessible web server at www.thegleelab.org/MLACP.html.
Collapse
Affiliation(s)
| | - Shaherin Basith
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sun Choi
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
186
|
Khandia R, Munjal A, Kumar A, Singh G, Karthik K, Dhama K. Cell Penetrating Peptides: Biomedical/Therapeutic Applications with Emphasis as Promising Futuristic Hope for Treating Cancer. INT J PHARMACOL 2017. [DOI: 10.3923/ijp.2017.677.689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
187
|
Xia X, Cheng L, Zhang S, Wang L, Hu J. The role of natural antimicrobial peptides during infection and chronic inflammation. Antonie van Leeuwenhoek 2017; 111:5-26. [PMID: 28856473 DOI: 10.1007/s10482-017-0929-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/15/2017] [Indexed: 01/12/2023]
Abstract
Natural antimicrobial peptides (AMPs), a family of small polypeptides that are produced by constitutive or inducible expression in organisms, are integral components of the host innate immune system. In addition to their broad-spectrum antibacterial activity, natural AMPs also have many biological activities against fungi, viruses and parasites. Natural AMPs exert multiple immunomodulatory roles that may predominate under physiological conditions where they lose their microbicidal properties in serum and tissue environments. Increased drug resistance among microorganisms is occurring far more quickly than the discovery of new antibiotics. Natural AMPs have shown promise as 'next generation antibiotics' due to their broad-spectrum curative effects, low toxicity, the fact that they are not residual in animals, and the low rates of resistance exhibited by many pathogens. Many types of synthetic AMPs are currently being tested in clinical trials for the prevention and treatment of various diseases such as chemotherapy-associated infections, diabetic foot ulcers, catheter-related infections, and other conditions. Here, we provide an overview of the types and functions of natural AMPs and their role in combating microorganisms and different infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China
| | - Likun Cheng
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, 256600, People's Republic of China
| | - Shouping Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China.
| |
Collapse
|
188
|
Usmani SS, Bedi G, Samuel JS, Singh S, Kalra S, Kumar P, Ahuja AA, Sharma M, Gautam A, Raghava GPS. THPdb: Database of FDA-approved peptide and protein therapeutics. PLoS One 2017; 12:e0181748. [PMID: 28759605 PMCID: PMC5536290 DOI: 10.1371/journal.pone.0181748] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/06/2017] [Indexed: 11/20/2022] Open
Abstract
THPdb (http://crdd.osdd.net/raghava/thpdb/) is a manually curated repository of Food and Drug Administration (FDA) approved therapeutic peptides and proteins. The information in THPdb has been compiled from 985 research publications, 70 patents and other resources like DrugBank. The current version of the database holds a total of 852 entries, providing comprehensive information on 239 US-FDA approved therapeutic peptides and proteins and their 380 drug variants. The information on each peptide and protein includes their sequences, chemical properties, composition, disease area, mode of activity, physical appearance, category or pharmacological class, pharmacodynamics, route of administration, toxicity, target of activity, etc. In addition, we have annotated the structure of most of the protein and peptides. A number of user-friendly tools have been integrated to facilitate easy browsing and data analysis. To assist scientific community, a web interface and mobile App have also been developed.
Collapse
Affiliation(s)
| | - Gursimran Bedi
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Jesse S. Samuel
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sandeep Singh
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sourav Kalra
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Pawan Kumar
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Anjuman Arora Ahuja
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Meenu Sharma
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Ankur Gautam
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
- * E-mail: , (GPSR); (AG)
| | - Gajendra P. S. Raghava
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
- * E-mail: , (GPSR); (AG)
| |
Collapse
|
189
|
Yuan Y, Guo B, Hao L, Liu N, Lin Y, Guo W, Li X, Gu B. Doxorubicin-loaded environmentally friendly carbon dots as a novel drug delivery system for nucleus targeted cancer therapy. Colloids Surf B Biointerfaces 2017; 159:349-359. [PMID: 28806666 DOI: 10.1016/j.colsurfb.2017.07.030] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/26/2017] [Accepted: 07/08/2017] [Indexed: 02/05/2023]
Abstract
Chemotherapy is widely applied against various kinds of carcinoma. Generally, chemotherapeutic agents, such as Doxorubicin (DOX), Paclitaxel (PTX), 5-Fluorouracil (5-FU), Methotrexate (MTX), and Vinblastine (VLB) are combined with a view to maximizing their efficacy. Unfortunately, chemotherapeutics are indiscriminate and also kill normal healthy cells, resulting in serious side effects. This non-productive and destructive distribution of chemotherapeutics is regarded as one of the largest problems associated with chemotherapy. Recently, the application of carbon dots (CDs) in cancer therapy has attracted considerable attention due to their attractive properties, such as biocompatibility and low toxicity. We report herein on the fabrication of CD-DOX antitumor drug complexes, from the combination of CDs and DOX, with a view to providing a novel and efficient strategy for cancer treatment. CDs were synthesized by hydrothermal treatment of milk, a simple and environmentally friendly synthetic process. DOX was conjugated to the CDs through electrostatic interactions via the multiple surface CD functional groups. The CD-DOX complexes exhibited pH-dependent DOX release behavior. A cytotoxicity study demonstrated that the CDs were non-cytotoxic in the range of concentrations used. Compared to free DOX, the CD-DOX complexes were significantly more destructive to the adenoid cystic carcinoma cell line (ACC-2), but exhibited lower toxicity to a mouse fibroblast cell line (L929). Confocal microscopy and flow cytometry confirmed that CD-DOX complexes increased cancer therapy efficiency through the localization of a much higher quantity of drugs in the nuclei of tumor cells and induced a higher rate of apoptosis in ACC-2 cells, compared to DOX alone.
Collapse
Affiliation(s)
- Yifang Yuan
- Institution of Stomatology, The General Hospital of China LA, Beijing 100853, China.
| | - Bin Guo
- Institution of Stomatology, The General Hospital of China LA, Beijing 100853, China.
| | - Liying Hao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Na Liu
- Institution of Stomatology, The General Hospital of China LA, Beijing 100853, China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Wushuang Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xiaoguang Li
- Institution of Stomatology, The General Hospital of China LA, Beijing 100853, China.
| | - Bin Gu
- Institution of Stomatology, The General Hospital of China LA, Beijing 100853, China.
| |
Collapse
|
190
|
Darrigues E, Dantuluri V, Nima ZA, Vang-Dings KB, Griffin RJ, Biris AR, Ghosh A, Biris AS. Raman spectroscopy using plasmonic and carbon-based nanoparticles for cancer detection, diagnosis, and treatment guidance. Part 2: Treatment. Drug Metab Rev 2017; 49:253-283. [DOI: 10.1080/03602532.2017.1307387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Emilie Darrigues
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Vijayalakshmi Dantuluri
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Zeid A. Nima
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Kieng Bao Vang-Dings
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Robert J. Griffin
- Arkansas Nanomedicine Center, Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alexandru R. Biris
- National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | - Anindya Ghosh
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Alexandru S. Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, USA
| |
Collapse
|
191
|
Cicero AFG, Fogacci F, Colletti A. Potential role of bioactive peptides in prevention and treatment of chronic diseases: a narrative review. Br J Pharmacol 2017; 174:1378-1394. [PMID: 27572703 PMCID: PMC5429326 DOI: 10.1111/bph.13608] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 08/12/2016] [Accepted: 08/18/2016] [Indexed: 12/24/2022] Open
Abstract
In the past few years, increasing interest has been directed to bioactive peptides of animal and plant origin: in particular, researchers have focused their attention on their mechanisms of action and potential role in the prevention and treatment of cancer, cardiovascular and infective diseases. We have developed a search strategy to identify these studies in PubMed (January 1980 to May 2016); particularly those papers presenting comprehensive reviews or meta-analyses, plus in vitro and in vivo studies and clinical trials on those bioactive peptides that affect cardiovascular diseases, immunity or cancer, or have antioxidant, anti-inflammatory and antimicrobial effects. In this review we have mostly focused on evidence-based healthy properties of bioactive peptides from different sources. Bioactive peptides derived from fish, milk, meat and plants have demonstrated significant antihypertensive and lipid-lowering activity in clinical trials. Many bioactive peptides show selective cytotoxic activity against a wide range of cancer cell lines in vitro and in vivo, whereas others have immunomodulatory and antimicrobial effects. Furthermore, some peptides exert anti-inflammatory and antioxidant activity, which could aid in the prevention of chronic diseases. However, clinical evidence is at an early stage, and there is a need for solid pharmacokinetic data and for standardized extraction procedures. Further studies on animals and randomized clinical trials are required to confirm these effects, and enable these peptides to be used as preventive or therapeutic treatments. LINKED ARTICLES This article is part of a themed section on Principles of Pharmacological Research of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.11/issuetoc.
Collapse
Affiliation(s)
- Arrigo F G Cicero
- Atherosclerosis and Metabolic Diseases Research Center, Medicine and Surgery DeptartmentAlma Mater Studiorum, University of BolognaBolognaItaly
| | - Federica Fogacci
- Atherosclerosis and Metabolic Diseases Research Center, Medicine and Surgery DeptartmentAlma Mater Studiorum, University of BolognaBolognaItaly
| | - Alessandro Colletti
- Atherosclerosis and Metabolic Diseases Research Center, Medicine and Surgery DeptartmentAlma Mater Studiorum, University of BolognaBolognaItaly
| |
Collapse
|
192
|
PEGylated doxorubicin nanoparticles mediated by HN-1 peptide for targeted treatment of oral squamous cell carcinoma. Int J Pharm 2017; 525:21-31. [DOI: 10.1016/j.ijpharm.2017.04.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/03/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022]
|
193
|
Solid matrix-based lipid nanoplatforms as carriers for combinational therapeutics in cancer. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0337-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
194
|
Niescioruk A, Nieciecka D, Puszko AK, Królikowska A, Kosson P, Perret GY, Krysinski P, Misicka A. Physicochemical properties and in vitro cytotoxicity of iron oxide-based nanoparticles modified with antiangiogenic and antitumor peptide A7R. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2017; 19:160. [PMID: 28503085 PMCID: PMC5406482 DOI: 10.1007/s11051-017-3859-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/11/2017] [Indexed: 06/07/2023]
Abstract
Superparamagnetic iron oxide-based nanoparticles (SPIONs) are promising carriers as targeted drug delivery vehicles, because they can be guided to their target with the help of an external magnetic field. Functionalization of nanoparticles' surface with molecules, which bind with high affinity to receptors on target tissue significantly facilitates delivery of coated nanoparticles to their targeted site. Here, we demonstrate conjugation of an antiangiogenic and antitumor peptide ATWLPPR (A7R) to SPIONs modified with sebacic acid (SPIONs-SA). Successful conjugation was confirmed by various analytical techniques (FTIR, SERS, SEM-EDS, TEM, TGA). Cell cytotoxicity studies, against two cell lines (HUVEC and MDA-MB-231) indicated that SPIONs modified with A7R reduced HUVEC cell viability at concentrations higher than 0.01 mg Fe/mL, in comparison to cells that were exposed to either the nanoparticles modified with sebacic acid or A7R peptide solely, what might be partially caused by a process of internalization.
Collapse
Affiliation(s)
- Anna Niescioruk
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Dorota Nieciecka
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Anna K. Puszko
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Agata Królikowska
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Piotr Kosson
- Department of Neuropeptides, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Gerard Y. Perret
- Sorbonne Paris Cité, Université Paris 13, INSERM U1125, 74 rue Marcel Cachin, 93017 Bobigny, France
| | - Pawel Krysinski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Aleksandra Misicka
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
- Department of Neuropeptides, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| |
Collapse
|
195
|
Marqus S, Pirogova E, Piva TJ. Evaluation of the use of therapeutic peptides for cancer treatment. J Biomed Sci 2017; 24:21. [PMID: 28320393 PMCID: PMC5359827 DOI: 10.1186/s12929-017-0328-x] [Citation(s) in RCA: 319] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/14/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer along with cardiovascular disease are the main causes of death in the industrialised countries around the World. Conventional cancer treatments are losing their therapeutic uses due to drug resistance, lack of tumour selectivity and solubility and as such there is a need to develop new therapeutic agents. Therapeutic peptides are a promising and a novel approach to treat many diseases including cancer. They have several advantages over proteins or antibodies: as they are (a) easy to synthesise, (b) have a high target specificity and selectivity and (c) have low toxicity. Therapeutic peptides do have some significant drawbacks related to their stability and short half-life. In this review, strategies used to overcome peptide limitations and to enhance their therapeutic effect will be compared. The use of short cell permeable peptides that interfere and inhibit protein-protein interactions will also be evaluated.
Collapse
Affiliation(s)
- Susan Marqus
- School of Engineering, RMIT University, Bundoora, VIC 3083 Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Bundoora, VIC 3083 Australia
| | - Terrence J. Piva
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| |
Collapse
|
196
|
Li Y, Shan F, Chen J. Lipid raft-mediated miR-3908 inhibition of migration of breast cancer cell line MCF-7 by regulating the interactions between AdipoR1 and Flotillin-1. World J Surg Oncol 2017; 15:69. [PMID: 28327197 PMCID: PMC5361711 DOI: 10.1186/s12957-017-1120-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/14/2017] [Indexed: 02/01/2023] Open
Abstract
Background The mechanisms of lipid raft regulation by microRNAs in breast cancer are not fully understood. This work focused on the evaluation and identification of miR-3908, which may be a potential biomarker related to the migration of breast cancer cells, and elucidates lipid-raft-regulating cell migration in breast cancer. Methods To confirm the prediction that miR-3908 is matched with AdipoR1, we used 3’-UTR luciferase activity of AdipoR1 to assess this. Then, human breast cancer cell line MCF-7 was cultured in the absence or presence of the mimics or inhibitors of miR-3908, after which the biological functions of MCF-7 cells were analyzed. The protein expression of AdipoR1, AMPK, and SIRT-1 were examined. The interaction between AdipoR1 and Flotillin-1, or its effects on lipid rafts on regulating cell migration of MCF-7, was also investigated. Results AdipoR1 is a direct target of miR-3908. miR-3908 suppresses the expression of AdipoR1 and its downstream pathway genes, including AMPK, p-AMPK, and SIRT-1. miR-3908 enhances the process of breast cancer cell clonogenicity. miR-3908 exerts its effects on the proliferation and migration of MCF-7 cells, which are mediated by lipid rafts regulating AdipoR1’s ability to bind Flotillin-1. Conclusions miR-3908 is a crucial mediator of the migration process in breast cancer cells. Lipid rafts regulate the interactions between AdipoR1 and Flotillin-1 and then the migration process associated with miR-3908 in MCF-7 cells. Our findings suggest that targeting miR-3908 and the lipid raft, may be a promising strategy for the treatment and prevention of breast cancer.
Collapse
Affiliation(s)
- Yuan Li
- Department of Obstetrics and Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100048, China
| | - Fei Shan
- Department of Cardiac Surgery, Affiliated Hospital of Medical College of Yan'an University, Yan'an, Shanxi, 716000, China
| | - Jinglong Chen
- Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
197
|
Abstract
Protein aggregation is involved in many diseases. Often, a unique aggregation-prone sequence polymerizes to form regular fibrils. Many oncogenic mutants of the tumor suppressor p53 rapidly aggregate but form amorphous fibrils. A peptide surrounding Ile254 is proposed to be the aggregation-driving sequence in cells. We identified several different aggregating sites from limited proteolysis of harvested aggregates and effects of mutations on kinetics and products of aggregation. We present a model whereby the amorphous nature of the aggregates results from multisite branching of polymerization after slow unfolding of the protein, which may be a common feature of aggregation of large proteins. Greatly lowering the aggregation propensity of any one single site, including the site of Ile254, by mutation did not inhibit aggregation in vitro because aggregation could still occur via the other sites. Inhibition of an individual site is, accordingly, potentially unable to prevent aggregation in vivo. However, cancer cells are specifically killed by peptides designed to inhibit the Ile254 sequence and further aggregation-driving sequences that we have found. Consistent with our proposed mechanism of aggregation, we found that such peptides did not inhibit aggregation of mutant p53 in vitro. The cytotoxicity was not eliminated by knockdown of p53 in 2D cancer cell cultures. The peptides caused rapid cell death, much faster than usually expected for p53-mediated transcription-dependent apoptosis. There may also be non-p53 targets for those peptides in cancer cells, such as p63, or the peptides may alter other interactions of partly denatured p53 with receptors.
Collapse
|
198
|
Haque A, Faizi MSH, Rather JA, Khan MS. Next generation NIR fluorophores for tumor imaging and fluorescence-guided surgery: A review. Bioorg Med Chem 2017; 25:2017-2034. [PMID: 28284863 DOI: 10.1016/j.bmc.2017.02.061] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 12/11/2022]
Abstract
Cancer is a group of diseases responsible for the major causes of mortality and morbidity among people of all ages. Even though medical sciences have made enormous growth, complete treatment of this deadly disease is still a challenging task. Last few decades witnessed an impressive growth in the design and development of near infrared (NIR) fluorophores with and without recognition moieties for molecular recognitions, imaging and image guided surgeries. The present article reviews recently reported NIR emitting organic/inorganic fluorophores that targets and accumulates in organelle/organs specifically for molecular imaging of cancerous cells. Near infrared (NIR probe) with or without a tumor-targeting warhead have been considered and discussed for their applications in the field of cancer imaging. In addition, challenges persist in this area are also delineated in this review.
Collapse
Affiliation(s)
- Ashanul Haque
- Department of Chemistry, College of Sciences, Sultan Qaboos University, Muscat, Oman.
| | | | - Jahangir Ahmad Rather
- Department of Chemistry, College of Sciences, Sultan Qaboos University, Muscat, Oman
| | - Muhammad S Khan
- Department of Chemistry, College of Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
199
|
Akrami H, Safavi M, Mirjalili BF, Dehghani Ashkezari M, Dadfar F, Mohaghegh N, Emami S, Salehi F, Nadri H, Ardestani SK, Firoozpour L, Khoobi M, Foroumadi A. Facile synthesis and antiproliferative activity of 7H-benzo[7,8]chromeno[2,3-d]pyrimidin-8-amines. Eur J Med Chem 2017; 127:128-136. [DOI: 10.1016/j.ejmech.2016.12.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/02/2016] [Accepted: 12/18/2016] [Indexed: 12/15/2022]
|
200
|
Hahn EM, Estrada-Ortiz N, Han J, Ferreira VFC, Kapp TG, Correia JDG, Casini A, Kühn FE. Functionalization of Ruthenium(II) Terpyridine Complexes with Cyclic RGD Peptides To Target Integrin Receptors in Cancer Cells. Eur J Inorg Chem 2016. [DOI: 10.1002/ejic.201601094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Eva M. Hahn
- Molecular Catalysis, Catalysis Research Center and Department of Chemistry; Technische Universität München; Lichtenbergstr. 4 85747 Garching bei München Germany
- School of Chemistry; Cardiff University; Park Place CF103AT Cardiff United Kingdom
| | - Natalia Estrada-Ortiz
- Groningen Research Institute of Pharmacy; University of Groningen; Antonius Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Jiaying Han
- Groningen Research Institute of Pharmacy; University of Groningen; Antonius Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Vera F. C. Ferreira
- Centro de Ciências e Tecnologias Nucleares; Instituto Superior Técnico; Universidade de Lisboa, CTN; Estrada Nacional 10 (km 139.7) 2695-066 Bobadela LRS Portugal
| | - Tobias G. Kapp
- Institute for Advanced Study; Technische Universität München; Lichtenbergstr. 2a 85748 Garching Germany
| | - João D. G. Correia
- Centro de Ciências e Tecnologias Nucleares; Instituto Superior Técnico; Universidade de Lisboa, CTN; Estrada Nacional 10 (km 139.7) 2695-066 Bobadela LRS Portugal
| | - Angela Casini
- School of Chemistry; Cardiff University; Park Place CF103AT Cardiff United Kingdom
- Groningen Research Institute of Pharmacy; University of Groningen; Antonius Deusinglaan 1 9713 AV Groningen The Netherlands
- Institute for Advanced Study; Technische Universität München; Lichtenbergstr. 2a 85748 Garching Germany
| | - Fritz E. Kühn
- Molecular Catalysis, Catalysis Research Center and Department of Chemistry; Technische Universität München; Lichtenbergstr. 4 85747 Garching bei München Germany
| |
Collapse
|