1
|
Roshanak S, Yarabbi H, Shahidi F, Tabatabaei Yazdi F, Movaffagh J, Javadmanesh A. Effects of adding poly-histidine tag on stability, antimicrobial activity and safety of recombinant buforin I expressed in periplasmic space of Escherichia coli. Sci Rep 2023; 13:5508. [PMID: 37015983 PMCID: PMC10073254 DOI: 10.1038/s41598-023-32782-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/02/2023] [Indexed: 04/06/2023] Open
Abstract
The lack of cost-effective methods for producing antimicrobial peptides has made it impossible to use their high potential as a new and powerful class of antimicrobial agents. In recent years, extensive research has been conducted to decrease the cost of recombinant proteins production through microorganisms, transgenic animals, and plants. Well-known genetic and physiological characteristics, short-term proliferation, and ease of manipulation make E. coli expression system a valuable host for recombinant proteins production. Expression in periplasmic space is recommended to reduce the inherently destructive behavior of antimicrobial peptides against the expressing microorganism and to decline susceptibility to proteolytic degradation. In this study, a pET-based expression system was used to express buforin I at E. coli periplasmic space, and its antimicrobial, hemolytic, and cell toxicity activities as well as structural stability were evaluated. The hemolysis activity and cytotoxicity of His-tagged buforin I were negligible and its antimicrobial activity did not show a significant difference compared to synthetic buforin I. In addition, in silico investigating of stability of native and His-tagged buforin I showed that RMSF, RMSD and Rg curves had followed a similar trend during 150 ns simulation. Furthermore, evaluating the modelled structures, FTIR and X-ray methods of both peptides indicated an insignificant structural difference. It was concluded that the recombinant buforin I could be a viable alternative to some currently used antibiotics by successfully expressing it in the pET-based expression system.
Collapse
Affiliation(s)
- Sahar Roshanak
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hanieh Yarabbi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fakhri Shahidi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Farideh Tabatabaei Yazdi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Jebraeil Movaffagh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Javadmanesh
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974, Razavi Khorasan Province, Iran.
- Industrial Biotechnology Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
2
|
Vakili B, Jahanian-Najafabadi A. Application of Antimicrobial Peptides in the Design and Production of Anticancer Agents. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10501-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
3
|
Kordi M, Borzouyi Z, Chitsaz S, Asmaei MH, Salami R, Tabarzad M. Antimicrobial peptides with anticancer activity: Today status, trends and their computational design. Arch Biochem Biophys 2023; 733:109484. [PMID: 36473507 DOI: 10.1016/j.abb.2022.109484] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Some antimicrobial peptides have been shown to be able to inhibit the proliferation of cancer cell lines. Various strategies for treating cancers with active peptides have been pursued. According to the reports, anticancer peptides are important therapeutic peptides, which can act through two distinct pathways: they either just create pores in the cell membrane, or they have a vital intracellular target. In this review, publications up to Sep. 2021 had extracted form Scopus and PubMed using "antimicrobial peptide" and "anticancer peptide" as keywords. In second step, "computational design" related publications extracted. Among publications, those have similar scopes were classified and selected based on mechanisms of action and application. In this review, the most recent advances in the field of antimicrobial peptides with anti-cancer activities have been summarized. Freely available webservers such as AntiCP, ACPP, iACP, iACP-GAEnsC, ACPred are discussed here. In conclusion, despite some limitations of ACPs such as production cost and challenges, short half-life and toxicity on normal cells, the beneficial properties of AMPs make some of them good therapeutic agents for cancer therapy. Towards designing novel ACPs, the computational methods have substantial position and have been used progressively, today.
Collapse
Affiliation(s)
- Masoumeh Kordi
- Department of Plant Science and Biotechnology, School of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Zeynab Borzouyi
- Department of Agriculture, School of Agriculture and Plant Breeding, Islamic Azad University, Sabzevar, Iran
| | - Saideh Chitsaz
- Department of Microbiology, Islamic Azad University, Karaj, Iran
| | | | - Robab Salami
- Department of Plant Science and Biotechnology, School of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Science, Iran.
| |
Collapse
|
4
|
Chowdary B N, Preetham HD, Verma SK, Hamse VK, Umashankara M, Raj. S N, Pramoda K, Kumar KSS, Selvi G. A short hydrophobic peptide conjugated 3,5- disubstituted pyrazoles as antibacterial agents with DNA gyrase inhibition. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
5
|
Kaewjanthong P, Sooksai S, Sasano H, Hutvagner G, Bajan S, McGowan E, Boonyaratanakornkit V. Cell-penetrating peptides containing the progesterone receptor polyproline domain inhibits EGF signaling and cell proliferation in lung cancer cells. PLoS One 2022; 17:e0264717. [PMID: 35235599 PMCID: PMC8890653 DOI: 10.1371/journal.pone.0264717] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/15/2022] [Indexed: 01/10/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for the majority (80-85%) of all lung cancers. All current available treatments have limited efficacy. The epidermal growth factor receptor (EGFR) plays a critical role in the development and progression of NSCLC, with high EGFR expression associated with increased cell proliferation and poor prognosis. Thus, interfering with EGFR signaling has been shown to effectively reduce cell proliferation and help in the treatment of NSCLC. We previously demonstrated that the progesterone receptor (PR) contains a polyproline domain (PPD) that directly interacts with Src homology 3 (SH3) domain-containing molecules and expression of PR-PPD peptides inhibits NSCLC cell proliferation. In this study, we investigated whether the introduction of PR-PPD by cell-penetrating peptides (CPPs) could inhibit EGF-induced cell proliferation in NSCLC cells. PR-PPD was attached to a cancer-specific CPP, Buforin2 (BR2), to help deliver the PR-PPD into NSCLC cells. Interestingly, addition of BR2-2xPPD peptides containing two PR-PPD repeats was more effective in inhibiting NSCLC proliferation and significantly reduced EGF-induced phosphorylation of Erk1/2. BR2-2xPPD treatment induced cell cycle arrest by inhibiting the expression of cyclin D1 and CDK2 genes in EGFR-wild type A549 cells. Furthermore, the combination treatment of EGFR-tyrosine kinase inhibitors (TKIs), including Gefitinib or Erlotinib, with BR2-2xPPD peptides further suppressed the growth of NSCLC PC9 cells harboring EGFR mutations as compared to EGFR-TKIs treatment alone. Importantly, BR2-2xPPD peptides mediated growth inhibition in acquired Gefitinib- and Erlotinib- resistant lung adenocarcinoma cells. Our data suggests that PR-PPD is the minimal protein domain sufficient to inhibit NSCLC cell growth and has the potential to be developed as a novel NSCLC therapeutic agent.
Collapse
Affiliation(s)
- Panthita Kaewjanthong
- Department of Clinical Chemistry and Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sarintip Sooksai
- The Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, Bangkok, Thailand
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Gyorgy Hutvagner
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Australia
| | - Sarah Bajan
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Australia
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Australia
- Sunshine Coast Health Institute, Birtinya, Australia
| | - Eileen McGowan
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Viroj Boonyaratanakornkit
- Department of Clinical Chemistry and Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
- Age-related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Silva ARP, Guimarães M, Rabelo J, Belen L, Perecin C, Farias J, Picado Madalena Santos JH, Rangel-Yagui CO. Recent advances in the design of antimicrobial peptide conjugates. J Mater Chem B 2022; 10:3587-3600. [DOI: 10.1039/d1tb02757c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antimicrobial peptides (AMPs) are ubiquitous host defense peptides characterized by antibiotic activity and lower propensity for developing resistance compared to classic antibiotics. While several AMPs have shown activity against antibiotic-sensitive...
Collapse
|
7
|
Roshanak S, Shahidi F, Tabatabaei Yazdi F, Javadmanesh A, Movaffagh J. Buforin I an alternative to conventional antibiotics: Evaluation of the antimicrobial properties, stability, and safety. Microb Pathog 2021; 161:105301. [PMID: 34822969 DOI: 10.1016/j.micpath.2021.105301] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/28/2022]
Abstract
Cationic antimicrobial peptides are being developed as a promising class of antimicrobial sub-stances. The introduction of a new antibiotic component requires a comprehensive study of its properties so that it can be relied upon to continue laboratory procedures and clinical trials on laboratory animals or human volunteers. Antimicrobial activity of buforin I was evaluated against 15 of the most important pathogenic bacterial and fungal strains. This was followed by assessing anti-biofilm activity, time-dependent inhibitory, thermal stability, plas-ma stability, hemolysis, and cytotoxic activities. The range of obtained MICs was between 4 and 16 μg/mL. The most resistant and most sensitive microbial strains were S. salivarius and C. perfringens, respectively. Buforin I not only inhibited biofilm formation, but also showed a high biofilm radiation activity. Buforin I was stable in human plasma and also at different temperatures including 40, 60, and 80 °C. Although no significant anti-cancer properties were observed for buforin I, the lack of cytotoxicity as well as the lack of hemolytic activity confirm its safety. The high therapeutic index indicated that buforin I has a considerable pharmaceutical potential and can be a reasonable candidate to replace antibiotics or administered in combination with antibiotics to increase the effectiveness as well as reduce the dose of antibiotics.
Collapse
Affiliation(s)
- Sahar Roshanak
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Fakhri Shahidi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Farideh Tabatabaei Yazdi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Ali Javadmanesh
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Jebraeil Movaffagh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Ciobanasu C. Peptides-based therapy and diagnosis. Strategies for non-invasive therapies in cancer. J Drug Target 2021; 29:1063-1079. [PMID: 33775187 DOI: 10.1080/1061186x.2021.1906885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, remarkable progress was registered in the field of cancer research. Though, cancer still represents a major cause of death and cancer metastasis a problem seeking for urgent solutions as it is the main reason for therapeutic failure. Unfortunately, the most common chemotherapeutic agents are non-selective and can damage healthy tissues and cause side effects that affect dramatically the quality of life of the patients. Targeted therapy with molecules that act specifically at the tumour sites interacting with overexpressed cancer receptors is a very promising strategy for achieving the specific delivery of anticancer drugs, radioisotopes or imaging agents. This review aims to give an overview on different strategies for targeting cancer cell receptors localised either at the extracellular matrix or at the cell membrane. Molecules like antibodies, aptamers and peptides targeting the cell surface are presented with advantages and disadvantages, with emphasis on peptides. The most representative peptides are described, including cell penetrating peptides, homing and anticancer peptides with particular consideration on recent discoveries.
Collapse
Affiliation(s)
- Corina Ciobanasu
- Sciences Department, Institute for Interdisciplinary Research, Alexandru I. Cuza University, Iaşi, Romania
| |
Collapse
|
9
|
Rastegari M, Shiri A, Behzad-Behbahani A, Rasoolian M, Zare F, Rafiei G, Mortazavi M, Sharifzadeh S, Hosseini SY. The Evaluation of tLyP-1-Bound Mda-7/IL-24 Killing Activity on a Liver Tumor Cell Line. Cancer Biother Radiopharm 2020; 36:827-836. [PMID: 32493109 DOI: 10.1089/cbr.2019.3080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Introduction: The melanoma differentiation-associated gene-7 (Mda-7)/interleukin-24 (IL-24) is a tumor killing cytokine, the bystander effect of which can be enhanced through tethering to tumor homing peptides (THPs). Materials and Methods: After fusing tLyP-1, RGR, and buforin as THPs to Mda-7/IL-24, enzyme-linked immunosorbent assay (ELISA) was used to determine the secretion potency of the recombinant proteins. The killing potency of plasmids expressing IL-24, IL-24.tLyP1, IL-24.RGR, and buf.IL-24 were assessed, using MTT, Annexin/PI staining assays as well as measuring the expression level of GADD-153 and BCL2-associated X (BAX) on Huh-7 cells. Three-dimensional structural analysis and protein-receptor interaction were also evaluated by modeling. Results: The ELISA result showed that contrary to IL-24.RGR and buf.IL-24, IL-24.tLyP-1 retained the secretion potency, similar to the native form. The viability assessments showed that IL-24 and IL-24.tLyP-1 had the most growth suppressive effects in comparison with the control group (p < 0.0001). Furthermore, IL-24 and IL-24.tLyP-1 had the highest apoptotic activity and significant upregulatory effect on the GADD-153 and BAX genes (p < 0.0003). The modeling showed that peptide modifications left no detrimental effect on IL-24 attachment to the cognate receptor. Conclusion: IL-24 can tolerate tLyP-1 peptide modification by retaining its secretion potency. Tethering tLyP-1 to IL-24 can induce more apoptosis than its modified versions by RGR or buforin.
Collapse
Affiliation(s)
- Mahroo Rastegari
- Department of Medical Biotechnology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Shiri
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Behzad-Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Rasoolian
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farahnaz Zare
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Rafiei
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Mortazavi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Sedigheh Sharifzadeh
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Tan J, Tay J, Hedrick J, Yang YY. Synthetic macromolecules as therapeutics that overcome resistance in cancer and microbial infection. Biomaterials 2020; 252:120078. [PMID: 32417653 DOI: 10.1016/j.biomaterials.2020.120078] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
Synthetic macromolecular antimicrobials have shown efficacy in the treatment of multidrug resistant (MDR) pathogens. These synthetic macromolecules, inspired by Nature's antimicrobial peptides (AMPs), mitigate resistance by disrupting microbial cell membrane or targeting multiple intracellular proteins or genes. Unlike AMPs, these polymers are less prone to degradation by proteases and are easier to synthesize on a large scale. Recently, various studies have revealed that cancer cell membrane, like that of microbes, is negatively charged, and AMPs can be used as anticancer agents. Nevertheless, efforts in developing polymers as anticancer agents has remained limited. This review highlights the recent advancement in the development of synthetic biodegradable antimicrobial polymers (e.g. polycarbonates, polyesters and polypeptides) and anticancer macromolecules including peptides and polymers. Additionally, strategies to improve their in vivo bioavailability and selectivity towards bacteria and cancer cells are examined. Lastly, future perspectives, including use of artificial intelligence or machine learning, in the development of antimicrobial and anticancer macromolecules are discussed.
Collapse
Affiliation(s)
- Jason Tan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore; Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Joyce Tay
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore; Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - James Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, CA, 95120, United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore.
| |
Collapse
|
11
|
Panjeta A, Preet S. Anticancer potential of human intestinal defensin 5 against 1, 2-dimethylhydrazine dihydrochloride induced colon cancer: A therapeutic approach. Peptides 2020; 126:170263. [PMID: 31981594 DOI: 10.1016/j.peptides.2020.170263] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
The escalating predicament of multidrug resistant cancer cells and associated side effects of conventional chemotherapy necessitates the exploration of alternative anticancer therapies. The present study evaluated anticancer therapeutic potential of human defensin 5 (HD-5) against colon cancer. The in vivo anticancer efficacy of HD-5 against 1,2-dimethylhydrazine (DMH) induced colon cancer was elucidated in terms of tumor biostatistics, number of aberrant crypt foci (ACF), in situ apoptosis assay,changes in morphological as well as histological architecture of colon(s). The direct interaction of peptide was investigated by incubating peptide with normal and/or cancerous colonocytes followed by phase contrast, Hoechst 3342 and AO/PI staining as well as confocal microscopy. Changes in membrane dynamics were evaluated by MC 540 and N-NBD-PE staining. In vivo decrease(s) in tumor parameters, number of aberrant crypt foci along with marked increase in the rate of apoptosis was observed.H&E staining revealed neutrophils infiltration and restoration of normal architecture in treated colon(s) which was consistent with scanning electron microscopic observations. Furthermore, non-membranolytic mechanism was found to be acquired by peptide as it could traverse cell membrane gaining access to nucleus and cytoplasm thereby disintegrating cellular architecture. MC 540 and NBD-PE staining revealed that peptide could bind to cancerous cells by taking advantage of altered fluidity levels. Our results indicated that HD-5 exhibited strong cancer cell killing and does not affect normal host cells. The peptide can be exploited as promising option to combat developing menace of colon cancer and/or can at least be used as an adjunct to present day chemotherapies.
Collapse
Affiliation(s)
- Anshul Panjeta
- Department of Biophysics, Basic Medical Sciences, Block II, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Simran Preet
- Department of Biophysics, Basic Medical Sciences, Block II, Panjab University, Sector 25, Chandigarh, 160014, India.
| |
Collapse
|
12
|
Kunda NK. Antimicrobial peptides as novel therapeutics for non-small cell lung cancer. Drug Discov Today 2020; 25:238-247. [DOI: 10.1016/j.drudis.2019.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/07/2019] [Accepted: 11/24/2019] [Indexed: 01/02/2023]
|
13
|
Li D, Xu Y. Buforin IIb induced cell cycle arrest in liver cancer. Anim Cells Syst (Seoul) 2019; 23:176-183. [PMID: 31231581 PMCID: PMC6566800 DOI: 10.1080/19768354.2019.1595139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 01/27/2023] Open
Abstract
The inhibitory effect of buforin IIb on different types of cancer, although not liver cancer, has been demonstrated previously. The aim of the present study was to investigate the effects of buforin IIb on the progression of liver cancer. The human liver cancer cell line HepG2 was treated with purified buforin IIb and the cell activity was determined by MTT, colony formation and transwell assays. The protein expression levels of cyclin-dependent kinases (CDKs) and cyclins were analyzed by western blotting and immunofluorescent cell staining. A tumor growth model was constructed using nude mice, and buforin IIb treatment was administered. The levels of CDK2 and cyclin A in the tumor tissues were detected by western blotting. Buforin IIb treatment depressed cell viability and colony formation and induced apoptosis significantly, and 1.0 µM concentration of buforin IIb was found to be the optimal dosage. The cell cycle was arrested at the G2/M phase following buforin IIb treatment. CDK2 and cyclin A were downregulated by treatment of the cells with 1.0 µM buforin IIb for 24 h. Treatment with buforin IIb also inhibited the migration of liver cancer cells in vitro. Furthermore, 50 nmol buforin IIb injection suppressed HepG2 cell subcutaneous tumor growth in the nude mouse model. Similar to the in vitro results, buforin IIb injection reduced the expression of CDK2 and cyclin A in the tumor tissue. these results demonstrate that buforin IIb inhibited liver cancer cell growth via the regulation of CDK2 and cyclin A expression.
Collapse
Affiliation(s)
- Dan Li
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Yong Xu
- Blood Purification Center, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| |
Collapse
|
14
|
Kenny RG, Marmion CJ. Toward Multi-Targeted Platinum and Ruthenium Drugs-A New Paradigm in Cancer Drug Treatment Regimens? Chem Rev 2019; 119:1058-1137. [PMID: 30640441 DOI: 10.1021/acs.chemrev.8b00271] [Citation(s) in RCA: 406] [Impact Index Per Article: 81.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
While medicinal inorganic chemistry has been practised for over 5000 years, it was not until the late 1800s when Alfred Werner published his ground-breaking research on coordination chemistry that we began to truly understand the nature of the coordination bond and the structures and stereochemistries of metal complexes. We can now readily manipulate and fine-tune their properties. This had led to a multitude of complexes with wide-ranging biomedical applications. This review will focus on the use and potential of metal complexes as important therapeutic agents for the treatment of cancer. With major advances in technologies and a deeper understanding of the human genome, we are now in a strong position to more fully understand carcinogenesis at a molecular level. We can now also rationally design and develop drug molecules that can either selectively enhance or disrupt key biological processes and, in doing so, optimize their therapeutic potential. This has heralded a new era in drug design in which we are moving from a single- toward a multitargeted approach. This approach lies at the very heart of medicinal inorganic chemistry. In this review, we have endeavored to showcase how a "multitargeted" approach to drug design has led to new families of metallodrugs which may not only reduce systemic toxicities associated with modern day chemotherapeutics but also address resistance issues that are plaguing many chemotherapeutic regimens. We have focused our attention on metallodrugs incorporating platinum and ruthenium ions given that complexes containing these metal ions are already in clinical use or have advanced to clinical trials as anticancer agents. The "multitargeted" complexes described herein not only target DNA but also contain either vectors to enable them to target cancer cells selectively and/or moieties that target enzymes, peptides, and intracellular proteins. Multitargeted complexes which have been designed to target the mitochondria or complexes inspired by natural product activity are also described. A summary of advances in this field over the past decade or so will be provided.
Collapse
Affiliation(s)
- Reece G Kenny
- Centre for Synthesis and Chemical Biology, Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| | - Celine J Marmion
- Centre for Synthesis and Chemical Biology, Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| |
Collapse
|
15
|
Vizovišek M, Vidmar R, Drag M, Fonović M, Salvesen GS, Turk B. Protease Specificity: Towards In Vivo Imaging Applications and Biomarker Discovery. Trends Biochem Sci 2018; 43:829-844. [PMID: 30097385 DOI: 10.1016/j.tibs.2018.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/05/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023]
Abstract
Proteases are considered of major importance in biomedical research because of their crucial roles in health and disease. Their ability to hydrolyze their protein and peptide substrates at single or multiple sites, depending on their specificity, makes them unique among the enzymes. Understanding protease specificity is therefore crucial to understand their biology as well as to develop tools and drugs. Recent advancements in the fields of proteomics and chemical biology have improved our understanding of protease biology through extensive specificity profiling and identification of physiological protease substrates. There are growing efforts to transfer this knowledge into clinical modalities, but their success is often limited because of overlapping protease features, protease redundancy, and chemical tools lacking specificity. Herein, we discuss the current trends and challenges in protease research and how to exploit the growing information on protease specificities for understanding protease biology, as well as for development of selective substrates, cleavable linkers, and activity-based probes and for biomarker discovery.
Collapse
Affiliation(s)
- Matej Vizovišek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia; These authors contributed equally to this work
| | - Robert Vidmar
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia; These authors contributed equally to this work
| | - Marcin Drag
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marko Fonović
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Guy S Salvesen
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
16
|
Han X, An L, Yan D, Hiroshi M, Ding W, Zhang M, Xu G, Sun Y, Yuan G, Wang M, Zhao N, Sun J, Zhu X, Du P. Combined antitumor effects of P-5m octapeptide and 5-fluorouracil on a murine model of H22 hepatoma ascites. Exp Ther Med 2018; 16:1586-1592. [PMID: 30186375 PMCID: PMC6122418 DOI: 10.3892/etm.2018.6422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/10/2017] [Indexed: 11/05/2022] Open
Abstract
The present study has demonstrated that P-5m octapeptide (P-5m) has therapeutic potential in metastatic human hepatocarcinoma, possibly through the modulation of matrix metalloproteinase-2 expression. The purpose of the present study was to evaluate the antitumor effect of P-5m combined with 5-fluorouracil (5-Fu) on the treatment of hepatoma 22 (H22) hepatocarcinoma malignant ascites in a mouse model. The inhibitory effect on the growth of mouse ascites tumors was monitored by measuring body weight gain, survival time, ascites volume, numbers of tumor cells, DNA synthesis and peritoneal capillary permeability analysis. The present data revealed a significant reduction in ascites volume and cell count in mice that were treated with P-5m plus 5-Fu. Furthermore, the median survival time in mice in the combination group was prolonged compared with the disease control group. Moreover, a significant reduction in the total H22 ascites cell count in mice from the combination group was observed when compared with the disease control group. P-5m plus 5-Fu was able to induce the cell cycle arrest and inhibit the peritoneal capillary permeability of the mice. To conclude, the present study indicated that P-5m may have therapeutic potential in ascites caused by hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiao Han
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Liping An
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Dongmei Yan
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Matsuura Hiroshi
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga 5202192, Japan
| | - Weiguang Ding
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga 5202192, Japan
| | - Mengchuan Zhang
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Guangyu Xu
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Ying Sun
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Guangxin Yuan
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Manli Wang
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Nanxi Zhao
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Jingbo Sun
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| | - Xun Zhu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Peige Du
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Changchun, Jilin 132013, P.R. China
| |
Collapse
|
17
|
Parker JP, Devocelle M, Morgan MP, Marmion CJ. Derivatisation of buforin IIb, a cationic henicosapeptide, to afford its complexation to platinum(ii) resulting in a novel platinum(ii)-buforin IIb conjugate with anti-cancer activity. Dalton Trans 2018; 45:13038-41. [PMID: 27292799 DOI: 10.1039/c6dt01510g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein we report the synthesis of buforin IIb, its novel malonate derivative malBuf and its Pt(ii) complex cis-[Pt(NH3)2(malBuf-2H)]. We decided to harness the cell targeting, cell-penetrating and anti-proliferative effects of buforin IIb to help target a cytotoxic dose of a Pt DNA binding species, {Pt(NH3)2} to cancer cells whilst also delivering a peptide with potent anti-cancer properties. Preliminary in vitro data shows cis-[Pt(NH3)2(malBuf-2H)] to be more cytotoxic against the cisplatin resistant ovarian cancer cell line (A2780cisR) relative to buforin IIb, cisplatin and cis-[Pt(NH3)2(malonate)].
Collapse
Affiliation(s)
- J P Parker
- Centre for Synthesis and Chemical Biology, Department of Pharmaceutical & Medicinal Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland.
| | - M Devocelle
- Centre for Synthesis and Chemical Biology, Department of Pharmaceutical & Medicinal Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland.
| | - M P Morgan
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - C J Marmion
- Centre for Synthesis and Chemical Biology, Department of Pharmaceutical & Medicinal Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
18
|
Li S, Zhang Y, Wang J, Zhao Y, Ji T, Zhao X, Ding Y, Zhao X, Zhao R, Li F, Yang X, Liu S, Liu Z, Lai J, Whittaker AK, Anderson GJ, Wei J, Nie G. Nanoparticle-mediated local depletion of tumour-associated platelets disrupts vascular barriers and augments drug accumulation in tumours. Nat Biomed Eng 2017; 1:667-679. [PMID: 31015598 DOI: 10.1038/s41551-017-0115-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 06/08/2017] [Indexed: 11/09/2022]
Abstract
Limited intratumoural perfusion and nanoparticle retention remain major bottlenecks for the delivery of nanoparticle therapeutics into tumours. Here, we show that polymer-lipid-peptide nanoparticles delivering the antiplatelet antibody R300 and the chemotherapeutic agent doxorubicin can locally deplete tumour-associated platelets, thereby enhancing vascular permeability and augmenting the accumulation of the nanoparticles in tumours. R300 is specifically released in the tumour on cleavage of the lipid-peptide shell of the nanoparticles by matrix metalloprotease 2, which is commonly overexpressed in tumour vascular endothelia and stroma, thus facilitating vascular breaches that enhance tumour permeability. We also show that this strategy leads to substantial tumour regression and metastasis inhibition in mice.
Collapse
Affiliation(s)
- Suping Li
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yinlong Zhang
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Jing Wang
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ying Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Tianjiao Ji
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiao Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yanping Ding
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiaozheng Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ruifang Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Feng Li
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiao Yang
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Shaoli Liu
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Zhaofei Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jianhao Lai
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, Centre for Magnetic Resonance, Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Gregory J Anderson
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia
| | - Jingyan Wei
- College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Guangjun Nie
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
19
|
Sol A, Skvirsky Y, Blotnick E, Bachrach G, Muhlrad A. Actin and DNA Protect Histones from Degradation by Bacterial Proteases but Inhibit Their Antimicrobial Activity. Front Microbiol 2016; 7:1248. [PMID: 27555840 PMCID: PMC4977296 DOI: 10.3389/fmicb.2016.01248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022] Open
Abstract
Histones are small polycationic proteins located in the cell nucleus. Together, DNA and histones are integral constituents of the nucleosomes. Upon apoptosis, necrosis, and infection – induced cell death, histones are released from the cell. The extracellular histones have strong antimicrobial activity but are also cytotoxic and thought as mediators of cell death in sepsis. The antimicrobial activity of the cationic extracellular histones is inhibited by the polyanionic DNA and F-actin, which also become extracellular upon cell death. DNA and F-actin protect histones from degradation by the proteases of Pseudomonas aeruginosa and Porphyromonas gingivalis. However, though the integrity of the histones is protected, the activity of histones as antibacterial agents is lost. The inhibition of the histone’s antibacterial activity and their protection from proteolysis by DNA and F-actin indicate a tight electrostatic interaction between the positively charged histones and negatively charged DNA and F-actin, which may have physiological significance in maintaining the equilibrium between the beneficial antimicrobial activity of extracellular histones and their cytotoxic effects.
Collapse
Affiliation(s)
- Asaf Sol
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine Jerusalem, Israel
| | - Yaniv Skvirsky
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine Jerusalem, Israel
| | - Edna Blotnick
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, Hebrew University of Jerusalem Jerusalem, Israel
| | - Gilad Bachrach
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine Jerusalem, Israel
| | - Andras Muhlrad
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine Jerusalem, Israel
| |
Collapse
|
20
|
Kim H, Jang JH, Kim SC, Cho JH. Enhancement of the antimicrobial activity and selectivity of GNU7 against Gram-negative bacteria by fusion with LPS-targeting peptide. Peptides 2016; 82:60-66. [PMID: 27242337 DOI: 10.1016/j.peptides.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 01/18/2023]
Abstract
Antimicrobial peptides (AMPs) provide a potential source of new antimicrobial therapeutics for the treatment of multidrug-resistant pathogens. To develop Gram-negative selective AMPs that can inhibit the effects of lipopolysaccharide (LPS)-induced sepsis, we added various rationally designed LPS-targeting peptides [amino acids 28-34 of lactoferrin (Lf28-34), amino acids 84-99 of bactericidal/permeability increasing protein (BPI84-99), and de novo peptide (Syn)] to the potent AMP, GNU7 (RLLRPLLQLLKQKLR). Compared to our original starting peptide GNU7, hybrid peptides had an 8- to 32-fold improvement in antimicrobial activity against Gram-negative bacteria, such as Escherichia coli and Salmonella typhimurium. Among them, Syn-GNU7 showed the strongest LPS-binding and -neutralizing activities, thus allowing it to selectively eliminate Gram-negative bacteria from within mixed cultures. Our results suggest that LPS-targeting peptides would be useful to increase the antimicrobial activity and selectivity of other AMPs against Gram-negative bacteria.
Collapse
Affiliation(s)
- Hyun Kim
- Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea
| | - Ju Hye Jang
- Research Institute of Life Science, Gyeongsang National University, Jinju 52828, South Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Ju Hyun Cho
- Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea; Research Institute of Life Science, Gyeongsang National University, Jinju 52828, South Korea.
| |
Collapse
|
21
|
Reinhardt A, Neundorf I. Design and Application of Antimicrobial Peptide Conjugates. Int J Mol Sci 2016; 17:E701. [PMID: 27187357 PMCID: PMC4881524 DOI: 10.3390/ijms17050701] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/25/2016] [Accepted: 05/04/2016] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial peptides (AMPs) are an interesting class of antibiotics characterized by their unique antibiotic activity and lower propensity for developing resistance compared to common antibiotics. They belong to the class of membrane-active peptides and usually act selectively against bacteria, fungi and protozoans. AMPs, but also peptide conjugates containing AMPs, have come more and more into the focus of research during the last few years. Within this article, recent work on AMP conjugates is reviewed. Different aspects will be highlighted as a combination of AMPs with antibiotics or organometallic compounds aiming to increase antibacterial activity or target selectivity, conjugation with photosensitizers for improving photodynamic therapy (PDT) or the attachment to particles, to name only a few. Owing to the enormous resonance of antimicrobial conjugates in the literature so far, this research topic seems to be very attractive to different scientific fields, like medicine, biology, biochemistry or chemistry.
Collapse
Affiliation(s)
- Andre Reinhardt
- Department of Chemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47, D-50674 Cologne, Germany.
| | - Ines Neundorf
- Department of Chemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47, D-50674 Cologne, Germany.
| |
Collapse
|
22
|
Kelly GJ, Kia AFA, Hassan F, O'Grady S, Morgan MP, Creaven BS, McClean S, Harmey JH, Devocelle M. Polymeric prodrug combination to exploit the therapeutic potential of antimicrobial peptides against cancer cells. Org Biomol Chem 2016; 14:9278-9286. [DOI: 10.1039/c6ob01815g] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The first targeted anticancer polymeric prodrug candidates of antimicrobial peptides, intended for combination therapy with another polymeric prodrug of an approved antineoplastic agent (doxorubicin) are reported.
Collapse
Affiliation(s)
- G. J. Kelly
- Centre for Synthesis and Chemical Biology
- Department of Pharmaceutical and Medicinal Chemistry
- Royal College of Surgeons in Ireland
- Dublin 2
- Ireland
| | | | - F. Hassan
- Molecular and Cellular Therapeutics
- Royal College of Surgeons in Ireland
- Dublin 2
- Ireland
| | - S. O'Grady
- Molecular and Cellular Therapeutics
- Royal College of Surgeons in Ireland
- Dublin 2
- Ireland
| | - M. P. Morgan
- Molecular and Cellular Therapeutics
- Royal College of Surgeons in Ireland
- Dublin 2
- Ireland
| | - B. S. Creaven
- Department of Science
- Institute of Technology Tallaght
- Dublin 24
- Ireland
| | - S. McClean
- Centre of Microbial Host Interactions
- Institute of Technology Tallaght
- Dublin 24
- Ireland
| | - J. H. Harmey
- Molecular and Cellular Therapeutics
- Royal College of Surgeons in Ireland
- Dublin 2
- Ireland
| | - M. Devocelle
- Centre for Synthesis and Chemical Biology
- Department of Pharmaceutical and Medicinal Chemistry
- Royal College of Surgeons in Ireland
- Dublin 2
- Ireland
| |
Collapse
|
23
|
Jang JH, Kim YJ, Kim H, Kim SC, Cho JH. Buforin IIb induces endoplasmic reticulum stress-mediated apoptosis in HeLa cells. Peptides 2015; 69:144-9. [PMID: 25958204 DOI: 10.1016/j.peptides.2015.04.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/22/2015] [Accepted: 04/25/2015] [Indexed: 12/13/2022]
Abstract
Buforin IIb, a novel cell-penetrating anticancer peptide derived from histone H2A, has been reported to induce mitochondria-dependent apoptosis in tumor cells. However, increasing evidence suggests that endoplasmic reticulum and mitochondria cooperate to signal cell death. In this study, we investigated the mechanism of buforin IIb-induced apoptosis in human cervical carcinoma HeLa cells by focusing on ER stress-mediated mitochondrial membrane permeabilization. Two-dimensional PAGE coupled with MALDI-TOF and western blot analysis showed that buforin IIb treatment of HeLa cells resulted in upregulation of ER stress proteins. PBA (ER stress inhibitor) and BAPTA/AM (Ca(2+) chelator) pretreatment rescued viability of buforin IIb-treated cells through abolishing phosphorylation of SAPK/JNK and p38 MAPK. SP600125 (SAPK/JNK inhibitor) and SB203580 (p38 MAPK inhibitor) attenuated down-regulation of Bcl-xL/Bcl-2, mitochondrial translocation of Bax, and cytochrome c release from mitochondria. Taken together, our data suggest that the ER stress pathway has an important role in the buforin IIb-induced apoptosis in HeLa cells.
Collapse
Affiliation(s)
- Ju Hye Jang
- Department of Biology, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, South Korea
| | - Yu Jin Kim
- Department of Biology, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, South Korea
| | - Hyun Kim
- Department of Biology, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, South Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Ju Hyun Cho
- Department of Biology, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, South Korea.
| |
Collapse
|
24
|
Forde E, Devocelle M. Pro-moieties of antimicrobial peptide prodrugs. Molecules 2015; 20:1210-27. [PMID: 25591121 PMCID: PMC6272668 DOI: 10.3390/molecules20011210] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/08/2015] [Indexed: 01/18/2023] Open
Abstract
Antimicrobial peptides (AMPs) are a promising class of antimicrobial agents that have been garnering increasing attention as resistance renders many conventional antibiotics ineffective. Extensive research has resulted in a large library of highly-active AMPs. However, several issues serve as an impediment to their clinical development, not least the issue of host toxicity. An approach that may allow otherwise cytotoxic AMPs to be used is to deliver them as a prodrug, targeting antimicrobial activity and limiting toxic effects on the host. The varied library of AMPs is complemented by a selection of different possible pro-moieties, each with their own characteristics. This review deals with the different pro-moieties that have been used with AMPs and discusses the merits of each.
Collapse
Affiliation(s)
- Eanna Forde
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin 9, Ireland.
| | - Marc Devocelle
- Centre for Synthesis and Chemical Biology, Department of Pharmaceutical and Medicinal Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
25
|
Gaspar D, Veiga AS, Castanho MARB. From antimicrobial to anticancer peptides. A review. Front Microbiol 2013; 4:294. [PMID: 24101917 PMCID: PMC3787199 DOI: 10.3389/fmicb.2013.00294] [Citation(s) in RCA: 494] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/11/2013] [Indexed: 12/26/2022] Open
Abstract
Antimicrobial peptides (AMPs) are part of the innate immune defense mechanism of many organisms. Although AMPs have been essentially studied and developed as potential alternatives for fighting infectious diseases, their use as anticancer peptides (ACPs) in cancer therapy either alone or in combination with other conventional drugs has been regarded as a therapeutic strategy to explore. As human cancer remains a cause of high morbidity and mortality worldwide, an urgent need of new, selective, and more efficient drugs is evident. Even though ACPs are expected to be selective toward tumor cells without impairing the normal body physiological functions, the development of a selective ACP has been a challenge. It is not yet possible to predict antitumor activity based on ACPs structures. ACPs are unique molecules when compared to the actual chemotherapeutic arsenal available for cancer treatment and display a variety of modes of action which in some types of cancer seem to co-exist. Regardless the debate surrounding the definition of structure-activity relationships for ACPs, great effort has been invested in ACP design and the challenge of improving effective killing of tumor cells remains. As detailed studies on ACPs mechanisms of action are crucial for optimizing drug development, in this review we provide an overview of the literature concerning peptides' structure, modes of action, selectivity, and efficacy and also summarize some of the many ACPs studied and/or developed for targeting different solid and hematologic malignancies with special emphasis on the first group. Strategies described for drug development and for increasing peptide selectivity toward specific cells while reducing toxicity are also discussed.
Collapse
Affiliation(s)
- Diana Gaspar
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de LisboaLisbon, Portugal
| | | | | |
Collapse
|
26
|
De novo generation of short antimicrobial peptides with enhanced stability and cell specificity. J Antimicrob Chemother 2013; 69:121-32. [DOI: 10.1093/jac/dkt322] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
27
|
Naumenkova TV, Antonov MY, Nikolaev IN, Shaitan KV. Effect of Pro11Ala substitution on the structural and functional properties of antimicrobial peptide buforin 2. Biophysics (Nagoya-shi) 2012. [DOI: 10.1134/s0006350912060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
28
|
Jang SA, Kim H, Lee JY, Shin JR, Kim DJ, Cho JH, Kim SC. Mechanism of action and specificity of antimicrobial peptides designed based on buforin IIb. Peptides 2012; 34:283-9. [PMID: 22306477 DOI: 10.1016/j.peptides.2012.01.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 01/19/2012] [Accepted: 01/19/2012] [Indexed: 11/19/2022]
Abstract
Buforin IIb-a synthetic analog of buforin II that contains a proline hinge between the two α-helices and a model α-helical sequence at the C-terminus (3× RLLR)-is a potent cell-penetrating antimicrobial peptide. To develop novel antimicrobial peptides with enhanced activities and specificity/therapeutic index, we designed several analogs (Buf III analogs) by substitutions of amino acids in the proline hinge region and two α-helices of buforin IIb, and examined their antimicrobial activity and mechanism of action. The substitution of hydrophobic residues ([F(6)] and [V(8)]) in the proline hinge region with other hydrophobic residues ([W(6)] and [I(8)]) did not affect antimicrobial activity, while the substitution of the first four amino acids RAGL with a model α-helical sequence increased the antimicrobial activity up to 2-fold. Like buforin IIb, Buf III analogs penetrated the bacterial cell membranes without significantly permeabilizing them and were accumulated inside Escherichia coli. Buf III analogs were shown to bind DNA in vitro and the DNA binding affinity of the peptides correlated linearly with their antimicrobial potency. Among the Buf III analogs, the therapeutic index of Buf IIIb and IIIc (RVVRQWPIG[RVVR](3) and KLLKQWPIG[KLLK](3), respectively) were improved 7-fold compared to that of buforin IIb. These results indicate that Buf III analogs appear to be promising candidates for future development as novel antimicrobial agents.
Collapse
Affiliation(s)
- Su A Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
29
|
Han X, Yan DM, Zhao XF, Hiroshi M, Ding WG, Li P, Jiang S, Du BR, Du PG, Zhu X. GHGKHKNK octapeptide (P-5m) inhibits metastasis of HCCLM3 cell lines via regulation of MMP-2 expression in in vitro and in vivo studies. Molecules 2012; 17:1357-72. [PMID: 22395332 PMCID: PMC6268966 DOI: 10.3390/molecules17021357] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 01/19/2012] [Accepted: 01/27/2012] [Indexed: 02/02/2023] Open
Abstract
P-5m, an octapeptide derived from domain 5 of HKa, was initially found to inhibit the invasion and migration of melanoma cells. The high metastatic potential of melanoma cells was prevented by the HGK motif in the P-5m peptide in vitro and in an experimental lung metastasis model, suggesting that P-5m may play an important role in the regulation of tumor metastasis. The aim of this study was to measure the effect of P-5m on tumor metastasis of human hepatocarcinoma cell line (HCCLM3) in vitro and in vivo in a nude mouse model of hepatocellular carcinoma (HCC), and detect the mechanisms involved in P-5m-induced anti-metastasis. By gelatin zymography, matrix metallo-proteinases 2 (MMP-2) activity in HCCLM3 was dramatically diminished by P-5m peptide. In addition, the migration and metastasis of HCCLM3 cells was also inhibited by the peptide in vitro. In an orthotopic model of HCC in nude mice, P-5m treatment effectively reduced the lung metastasis as well as the expression of MMP-2 in the tumor tissues. Overall, these observations indicate an important role for P-5m peptide in HCC invasion and metastasis, at least partially through modulation MMP-2 expression. These data suggests that P-5m may have therapeutic potential in metastatic human hepatocarcinoma.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Biological Products/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Movement/genetics
- Disease Models, Animal
- Female
- Humans
- Kininogen, High-Molecular-Weight/genetics
- Kininogen, High-Molecular-Weight/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Matrix Metalloproteinase 2/biosynthesis
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Oligopeptides/pharmacology
Collapse
Affiliation(s)
- Xiao Han
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China; (X.H.); (D.-M.Y.); (X.-F.Z.); (P.L.); (B.-R.D.)
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Jilin 132001, China;
| | - Dong-Mei Yan
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China; (X.H.); (D.-M.Y.); (X.-F.Z.); (P.L.); (B.-R.D.)
| | - Xiang-Feng Zhao
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China; (X.H.); (D.-M.Y.); (X.-F.Z.); (P.L.); (B.-R.D.)
| | - Matsuura Hiroshi
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan; (M.H.); (W.-G.D.)
| | - Wei-Guang Ding
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan; (M.H.); (W.-G.D.)
| | - Peng Li
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China; (X.H.); (D.-M.Y.); (X.-F.Z.); (P.L.); (B.-R.D.)
| | - Shuang Jiang
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Jilin 132001, China;
| | - Bai-Rong Du
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China; (X.H.); (D.-M.Y.); (X.-F.Z.); (P.L.); (B.-R.D.)
| | - Pei-Ge Du
- Department of Microbial and Biochemical Pharmacy, College of Pharmaceutical Science, Beihua University, Jilin 132001, China;
- Authors to whom correspondence should be addressed; (P.-G.D.); (X.Z.); Tel.: +86-431-8561-9476
| | - Xun Zhu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China; (X.H.); (D.-M.Y.); (X.-F.Z.); (P.L.); (B.-R.D.)
- Authors to whom correspondence should be addressed; (P.-G.D.); (X.Z.); Tel.: +86-431-8561-9476
| |
Collapse
|