1
|
Abedi Tameh F, Mohamed HEA, Aghababaee L, Akbari M, Alikhah Asl S, Javadi MH, Aucamp M, Cloete KJ, Soleimannejad J, Maaza M. In-vitro cytotoxicity of biosynthesized nanoceria using Eucalyptus camaldulensis leaves extract against MCF-7 breast cancer cell line. Sci Rep 2024; 14:17465. [PMID: 39075175 PMCID: PMC11286930 DOI: 10.1038/s41598-024-68272-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024] Open
Abstract
Cerium oxide nanoparticles possess unique properties that make them promising candidates in various fields, including cancer treatment. Among the proposed synthesis methods for CNPs, biosynthesis using natural extracts, offers an eco-friendly and convenient approach for producing CNPs, particularly for biomedical applications. In this study, a novel method of biosynthesis using the aqueous extract of Eucalyptus camaldulensis leaves was used to synthesize CNPs. Scanning electron microscopy and Transmission electron microscopy (TEM) techniques revealed that the synthesized CNPs exhibit a flower-like morphology. The particle size of CNPs obtained using Powder X-ray diffraction peaks and TEM as 13.43 and 39.25 nm. Energy-dispersive X-ray spectroscopy and Fourier transform infrared spectroscopy confirmed the effect of biomolecules during the synthesis process and the formation of CNPs. The cytotoxicity of biosynthesized samples was evaluated using the MTT method demonstrating the potential of these samples to inhibit MCF-7 cancerous cells. The viability of the MCF-7 cell line conducted by live/dead imaging assay confirmed the MTT cytotoxicity method and indicated their potential to inhibit cancerous cells. Furthermore, the successful uptake of CNPs by MCF-7 cancer cells, as demonstrated by confocal microscopy, provides evidence that the intracellular pathway contributes to the anticancer activity of the CNPs. In general, results indicate that the biosynthesized CNPs exhibit significant cytotoxicity against the MCF-7 cancerous cell line, attributed to their high surface area.
Collapse
Affiliation(s)
- Fatemeh Abedi Tameh
- UNESCO-UNISA-iTLABS Africa Chair in Nanoscience and Nanotechnology, College of Graduate Studies, University of South Africa, Muckleneuk Ridge, P.O. Box 392, Pretoria, 0003, South Africa.
- School of Chemistry, College of Science, University of Tehran, P.O. Box 141556455, Tehran, Iran.
| | - Hamza Elsayed Ahmed Mohamed
- UNESCO-UNISA-iTLABS Africa Chair in Nanoscience and Nanotechnology, College of Graduate Studies, University of South Africa, Muckleneuk Ridge, P.O. Box 392, Pretoria, 0003, South Africa
| | - Leila Aghababaee
- Neuroscience Laboratory, Institute of Biochemistry and Biophysics (IBB), Bio Organic, University of Tehran, Tehran, 1417614335, Iran
| | - Mahmood Akbari
- UNESCO-UNISA-iTLABS Africa Chair in Nanoscience and Nanotechnology, College of Graduate Studies, University of South Africa, Muckleneuk Ridge, P.O. Box 392, Pretoria, 0003, South Africa.
| | - Shervin Alikhah Asl
- UNESCO-UNISA-iTLABS Africa Chair in Nanoscience and Nanotechnology, College of Graduate Studies, University of South Africa, Muckleneuk Ridge, P.O. Box 392, Pretoria, 0003, South Africa
| | - Mohammad Hasan Javadi
- Department of Chemistry, Sharif University of Technology, P.O. Box 11155‑9516, Tehran, Iran
| | - Marique Aucamp
- School of Pharmacy, University of the Western Cape, Robert Sobukwe Drive, Bellville, 7130, Cape Town, South Africa
| | - Karen Jacqueline Cloete
- UNESCO-UNISA-iTLABS Africa Chair in Nanoscience and Nanotechnology, College of Graduate Studies, University of South Africa, Muckleneuk Ridge, P.O. Box 392, Pretoria, 0003, South Africa
| | - Janet Soleimannejad
- School of Chemistry, College of Science, University of Tehran, P.O. Box 141556455, Tehran, Iran
| | - Malik Maaza
- UNESCO-UNISA-iTLABS Africa Chair in Nanoscience and Nanotechnology, College of Graduate Studies, University of South Africa, Muckleneuk Ridge, P.O. Box 392, Pretoria, 0003, South Africa
| |
Collapse
|
2
|
Kong R, Wang N, Zhou C, Zhou Y, Guo X, Wang D, Shi Y, Wan R, Zheng Y, Lu J. Sanguinarine Induces Necroptosis of HCC by Targeting PKM2 Mediated Energy Metabolism. Cancers (Basel) 2024; 16:2533. [PMID: 39061173 PMCID: PMC11274805 DOI: 10.3390/cancers16142533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUNDS Abnormal metabolism is the hallmark of hepatocellular carcinoma. Targeting energy metabolism has become the major focus of cancer therapy. The natural product, sanguinarine, displays remarkable anti-tumor properties by disturbing energy homeostasis; however, the underlying mechanism has not yet been elucidated. METHODS The anticancer activity of sanguinarine was determined using CCK-8 and colony formation assay. Morphological changes of induced cell death were observed under electron microscopy. Necroptosis and apoptosis related markers were detected using western blotting. PKM2 was identified as the target by transcriptome sequencing. Molecular docking assay was used to evaluate the binding affinity of sanguinarine to the PKM2 molecule. Furthermore, Alb-CreERT2; PKM2loxp/loxp; Rosa26RFP mice was used to construct the model of HCC-through the intervention of sanguinarine in vitro and in vivo-to accurately explore the regulation effect of sanguinarine on cancer energy metabolism. RESULTS Sanguinarine inhibited tumor proliferation, metastasis and induced two modes of cell death. Molecular docking of sanguinarine with PKM2 showed appreciable binding affinity. PKM2 kinase activity and aerobic glycolysis rate declined, and mitochondrial oxidative phosphorylation was inhibited by sanguinarine application; these changes result in energy deficits and lead to necroptosis. Additionally, sanguinarine treatment prevents the translocation of PKM2 into the nucleus and suppresses the interaction of PKM2 with β-catenin; the transcriptional activity of PKM2/β-catenin signaling and its downstream genes were decreased. CONCLUSIONS Sanguinarine showed remarkable anti-HCC activity via regulating energy metabolism by PKM2/β-catenin signaling. On the basis of these investigations, we propose that sanguinarine might be considered as a promising compound for discovery of anti-HCC drugs.
Collapse
Affiliation(s)
- Rui Kong
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Nan Wang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200072, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China;
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Yuqing Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Xiaoyan Guo
- Department of Gastroenterology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
| | - Dongyan Wang
- Department of Gastroenterology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
| | - Yihai Shi
- Department of Gastroenterology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China;
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China;
- Department of Gastroenterology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
| |
Collapse
|
3
|
Ji K, Yao Y, Gao Y, Huang S, Ma L, Pan Q, Wu J, Zhang W, Chen H, Zhang L. Evaluating the cytotoxicity mechanism of the cell-penetrating peptide TP10 on Jurkat cells. Biochimie 2024; 221:182-192. [PMID: 37922978 DOI: 10.1016/j.biochi.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
TP10, a classic cell-penetrating peptide, shows a high degree of similarity to AMPs in structure. Although TP10 has been widely used in drug delivery, the mechanism underlying its cytotoxicity is yet to be elucidated. Herein, we explored the cell-killing mechanism of TP10 against human leukemia Jurkat cells. TP10 induced necrosis in Jurkat cells via rapid disruption of cell membranes, particularly at high concentrations. Although mitochondria in Jurkat cells were damaged by TP10, mitochondria-mediated apoptosis did not occur, possibly due to intracellular ATP depletion. Necroptosis in TP10-treated Jurkat cells became an alternative route of apoptosis. Our results demonstrate that necrosis and necroptosis rather than apoptosis are involved in the cell-killing mechanism of TP10, which contributes to the understanding of its toxicity.
Collapse
Affiliation(s)
- Kun Ji
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Yufan Yao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yuxuan Gao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Sujie Huang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Ling Ma
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qing Pan
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Jun Wu
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Wei Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, 730000, China.
| | - Hongmei Chen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Lei Zhang
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
4
|
McFetridge ML, Kulkarni K, Lee TH, Del Borgo MP, Aguilar MI, Ricardo SD. Elucidating the cell penetrating properties of self-assembling β-peptides. NANOSCALE 2023; 15:14971-14980. [PMID: 37661822 DOI: 10.1039/d3nr03673a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Self-assembling lipopeptide hydrogels have been widely developed for the delivery of therapeutics due to their rapid gelation, injectability, and highly controlled physicochemical properties. Lipopeptides are also known for their membrane-associating and cell penetrating properties, which may impact on their application in cell-encapsulation. Self-assembling lipidated-β3-peptide materials developed in our laboratory have previously been used in cell culture as 2D substrates, thus as a continuation of this work we aimed to encapsulate cells in 3D by forming a hydrogel. We therefore assessed the self-assembling lipidated-β3-peptides for cell-penetrating properties in mesenchymal stems cells (MSC) using fluorescence microscopy and membrane association with surface plasmon resonance spectroscopy (SPR). The results demonstrated that lipidated β3-peptides penetrate the MSC plasma membrane and localise to the mitochondrial network. While self-assembling lipopeptide hydrogels have shown tremendous potential for delivery of therapeutics, further optimisation may be required to minimise the membrane uptake of the lipidated-β3-peptides for cell encapsulation applications.
Collapse
Affiliation(s)
- Meg L McFetridge
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Mark P Del Borgo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
5
|
Tkachenko A, Onishchenko A, Myasoedov V, Yefimova S, Havranek O. Assessing regulated cell death modalities as an efficient tool for in vitro nanotoxicity screening: a review. Nanotoxicology 2023; 17:218-248. [PMID: 37083543 DOI: 10.1080/17435390.2023.2203239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Nanomedicine is a fast-growing field of nanotechnology. One of the major obstacles for a wider use of nanomaterials for medical application is the lack of standardized toxicity screening protocols for assessing the safety of newly synthesized nanomaterials. In this review, we focus on less frequently studied nanomaterials-induced regulated cell death (RCD) modalities, including eryptosis, necroptosis, pyroptosis, and ferroptosis, as a tool for in vitro nanomaterials safety evaluation. We summarize the latest insights into the mechanisms that mediate these RCDs in response to nanomaterials exposure. Comprehensive data from reviewed studies suggest that ROS (reactive oxygen species) overproduction and ROS-mediated pathways play a central role in nanomaterials-induced RCDs activation. On the other hand, studies also suggest that individual properties of nanomaterials, including size, shape, or surface charge, could determine specific toxicity pathways with consequent RCD induction as well. We anticipate that the evaluation of RCDs can become one of the mechanism-based screening methods in nanotoxicology. In addition to the toxicity assessment, evaluation of necroptosis-, pyroptosis-, and ferroptosis-promoting capacity of nanomaterials could simultaneously provide useful information for specific medical applications as could be their anti-tumor potential. Moreover, a detailed understanding of molecular mechanisms driving nanomaterials-mediated induction of immunogenic RCDs will substantially aid novel anti-tumor nanodrugs development.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Anatolii Onishchenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Valeriy Myasoedov
- Department of Medical Biology, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Svetlana Yefimova
- Institute for Scintillation Materials, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
6
|
In Silico Prediction of Anti-Infective and Cell-Penetrating Peptides from Thalassophryne nattereri Natterin Toxins. Pharmaceuticals (Basel) 2022; 15:ph15091141. [PMID: 36145362 PMCID: PMC9501638 DOI: 10.3390/ph15091141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 12/14/2022] Open
Abstract
The therapeutic potential of venom-derived peptides, such as bioactive peptides (BAPs), is determined by specificity, stability, and pharmacokinetics properties. BAPs, including anti-infective or antimicrobial peptides (AMPs) and cell-penetrating peptides (CPPs), share several physicochemical characteristics and are potential alternatives to antibiotic-based therapies and drug delivery systems, respectively. This study used in silico methods to predict AMPs and CPPs derived from natterins from the venomous fish Thalassophryne nattereri. Fifty-seven BAPs (19 AMPs, 8 CPPs, and 30 AMPs/CPPs) were identified using the web servers CAMP, AMPA, AmpGram, C2Pred, and CellPPD. The physicochemical properties were analyzed using ProtParam, PepCalc, and DispHred tools. The membrane-binding potential and cellular location of each peptide were analyzed using the Boman index by APD3, and TMHMM web servers. All CPPs and two AMPs showed high membrane-binding potential. Fifty-four peptides were located in the plasma membrane. Peptide immunogenicity, toxicity, allergenicity, and ADMET parameters were evaluated using several web servers. Sixteen antiviral peptides and 37 anticancer peptides were predicted using the web servers Meta-iAVP and ACPred. Secondary structures and helical wheel projections were predicted using the PEP-FOLD3 and Heliquest web servers. Fifteen peptides are potential lead compounds and were selected to be further synthesized and tested experimentally in vitro to validate the in silico screening. The use of computer-aided design for predicting peptide structure and activity is fast and cost-effective and facilitates the design of potent therapeutic peptides. The results demonstrate that toxins form a natural biotechnological platform in drug discovery, and the presence of CPP and AMP sequences in toxin families opens new possibilities in toxin biochemistry research.
Collapse
|
7
|
Stachowicz K. Is PSD-95 entangled in the side effects of antidepressants? Neurochem Int 2022; 159:105391. [PMID: 35817245 DOI: 10.1016/j.neuint.2022.105391] [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: 05/26/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 01/13/2023]
Abstract
PSD-95 is a component and a building block of an excitatory synapse. PSD-95 is a specialized protein that is part of a "combination lock" system responsible for plastic events at the synapse, such as receptor expression, which consequently induces changes in the PSD structure and thus affects synaptic plasticity. The possible involvement of PSD-95 in antidepressant side effects related to cognitive function and psychosis will be considered. An attempt will be made to trace the sequence of events in the proposed mechanism leading to these disorders, focusing mainly on NMDA receptors. Understanding the mechanisms of action of compounds with antidepressant potential may facilitate the design of safer drugs.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna, 12, 31-343, Kraków, Poland.
| |
Collapse
|
8
|
Marzhoseyni Z, Shayestehpour M, Salimian M, Esmaeili D, Saffari M, Fathizadeh H. Designing a novel fusion protein from Streptococcus agalactiae with apoptosis induction effects on cervical cancer cells. Microb Pathog 2022; 169:105670. [PMID: 35809755 DOI: 10.1016/j.micpath.2022.105670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022]
Abstract
Cervical cancer remains life-threatening cancer in women around the world. Due to the limitations of conventional treatment approaches, there is an urgent need to develop novel and more efficient strategies against cervical cancer. Therefore, the researchers attend to the alternative anti-cancer compounds like bacterial products. Rib and α are known as surface proteins of Streptococcus agalactiae with immunologic effects. In the present study, we designed a new anti-cancer fusion protein (Rib-α) originating from S. agalactiae with in silico methods, and then, the recombinant gene was cloned in the pET-22 (+) expression vector. The recombinant protein was expressed in E. coli BL21. To purify the expressed protein, we applied the Ni-NTA column. The molecular mechanism by which Rib-α is cytotoxic to cancer cells has been discussed based on MTT, flow cytometry, and real-time PCR methods. The engineered fusion protein suppressed the proliferation of the cancer cells at 180 μg/ml. Cytotoxic assessment and morphological changes, augmentation of apoptotic-related genes, upregulation of caspase-3 mRNA, and flow cytometric analysis confirmed that apoptosis might be the principal mechanism of cell death. According to our findings, Rib-α fusion protein motivated the intrinsic apoptosis pathway. Therefore, it can be an exciting candidate to discover a new class of antineoplastic agents.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Shayestehpour
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Morteza Salimian
- Anatomical Science Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Davoud Esmaeili
- Department of Microbiology and Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mahmood Saffari
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hadis Fathizadeh
- Student Research Committee, Sirjan School of Medical Sciences, Sirjan, Iran; Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| |
Collapse
|
9
|
Ma L, Huang S, Xie H, Ma P, Jia B, Yao Y, Gao Y, Li W, Song J, Zhang W. Influence of chain length on the anticancer activity of the antimicrobial peptide CAMEL with fatty acid modification. Eur J Med Chem 2022; 239:114557. [PMID: 35759906 DOI: 10.1016/j.ejmech.2022.114557] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 01/10/2023]
Abstract
Antimicrobial peptides (AMPs) display promising potential in cancer therapy. Modification with fatty acids is a simple and effective approach to improve the activity of AMPs. In the present study, we investigated the effects of fatty acid chain lengths on the anticancer activity, self-assembly and mechanism of action of CAMEL (CM15, KWKLFKKIGAVLKVL-NH2), an amphipathic AMP with 15 amino acids. Conjugation of fatty acids could obviously improve the in vitro anticancer activity of CAMEL. Among the tested peptides, C12-CAMEL showed the highest anticancer activity, while C16-CAMEL killed cancer cells with the slowest kinetics. This may be related to the self-assembly of C12-CAMEL and C16-CAMEL, which could form spherical nanoparticles and tightened nanofibers, respectively. In addition, necrosis and necroptosis rather than apoptosis were the major mechanisms underlying the anticancer activity of CAMEL, C12-CAMEL and C16-CAMEL, implying that modification with fatty acids did not obviously alter the mechanism of action of CAMEL. Notably, C12-CAMEL, with high and rapid cell-killing activity, exhibited significantly stronger in vivo anticancer activity than CAMEL and C16-CAMEL. Overall, the present work suggests that the choice of a suitable fatty acid for structural modification is necessary for improving the anticancer activity of AMPs.
Collapse
Affiliation(s)
- Ling Ma
- The Institute of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Sujie Huang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Huan Xie
- Department of Medical, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Panpan Ma
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Bo Jia
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yufan Yao
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yuxuan Gao
- The Institute of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Wenyuan Li
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jingjing Song
- The Institute of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Wei Zhang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
10
|
Behzadi M, Eghtedardoost M, Bagheri M. Endocytosis Involved d-Oligopeptide of Tryptophan and Arginine Displays Ordered Nanostructures and Cancer Cell Stereoselective Toxicity by Autophagy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14928-14943. [PMID: 35319877 DOI: 10.1021/acsami.1c23846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Owing to their self-aggregation propensity and selective interaction with the anionic membranes, the peptides rich in tryptophan (Trp) and arginine (Arg) are considered for the development of novel anticancer therapeutics. However, the structural insights from the perspective of backbone chirality and spatial orientation of side chains into the selective toxicity of peptides are limited. Here, we investigated the selectivity and cellular uptake of HHC36, a Trp/Arg-rich nonapeptide, and its d-enantiomer (allDHHC36) and a retroinverso analogue in the lung A549 and breast MDA-MB-231 cancer cells. We realized that the d-peptides can specifically induce autophagy at nontoxic concentrations only in the A549 cells supported from the LC 3-II immunostaining expression in the vicinity of the nucleus and the ultrastructural analysis revealing the autophagosome formation. The autophagic flux was also remarkable in the cells exposed to d-peptides at a far lower concentration in synergism with doxorubicin (DOX). In marked contrast, nonselective cell death was observed only if a high amount of HHC36 was applied. HHC36 tended to irregular collagen-like fibrils relative to allDHHC36 that distinctly formed higher-order coiled nanostructures. Interestingly, the short d-peptide fragments were generated in a harsh oxidative condition. Compared with the direct membrane transduction of HHC36, the entry of d-peptides into the lung cancer cells was controlled by endocytosis through the contribution of heparan sulfate proteoglycans (HSPGs) and cholesterol (CHO). However, both l- and d-peptides feasibly crossed the membrane and localized inside the S-phase-arrested cell nucleus. This suggested the likelihood of peptide intercalation with DNA that might differently appear in selective and/or nonselective deaths. These results unraveled the d-handedness-selective toxicity of a self-assembling Trp/Arg-rich sequence that is dependent on the cell type from the aspects of the density of anionic charges and CHO in the outer leaflet of the plasma membrane, as well as the intracellular redox imbalance that may drive the formation of toxic peptide nanostructure fragments.
Collapse
Affiliation(s)
- Malihe Behzadi
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335 Tehran, Iran
| | - Marzieh Eghtedardoost
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335 Tehran, Iran
| | - Mojtaba Bagheri
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335 Tehran, Iran
| |
Collapse
|
11
|
Bak IG, Chae CG, Lee JS. Synthetic Control of Helical Polyisocyanates by Living Anionic Polymerization toward Peptide Mimicry. Macromolecules 2022. [DOI: 10.1021/acs.macromol.1c02160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- In Gyu Bak
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Chang-Geun Chae
- Advanced Materials Division, Korea Research Institute of Chemical Technology (KRICT), 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Jae-Suk Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
12
|
Behzadi M, Amininasab M, Eghtedardoost M, Bagheri M. Turn-folded magainin lipopeptide analog induces cytoplasmic vacuoles in MDA-MB-231 cells through G2-phase arrest. Biochem Biophys Res Commun 2021; 583:199-205. [PMID: 34752987 DOI: 10.1016/j.bbrc.2021.10.076] [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] [Received: 09/12/2021] [Accepted: 10/30/2021] [Indexed: 02/04/2023]
Abstract
Selective induced non-canonical programmed deaths in the lipid raft type 1-enriched MDA-MB-231 is a promising treatment approach. Cationic amphiphilic peptides conjugated to relatively long fatty acyl chains that tend to self-aggregate are prone to upregulate necroptotic and paraptotic signaling. We investigated the toxic effects of an N-terminally palmitoylated magainin derivate (P1MK5E) in the MDA-MB-231 cells in relation to its structure at molecular level. The modeling showed that the palmitoylation reinforces a turn-like structural motif in the lipopeptide which is likely required for its activity. P1MK5E triggered intracellular generation of reactive oxygen species (ROS), G2-phase arrest, mitochondrial membrane potential (ΔΨmt) disturbance and presumable flopping of phosphatidylserine (PtdSer) to the cancer cell membrane outer surface in a comparable manner to doxorubicin (DOX) that induces apoptotic signaling. Despite forming extensive congregates of different sizes at the cell surface, P1MK5E had little impacts on the MDA-MB-231 membrane integrity. The cell death upon exposure to the lipopeptide was, however, caspase 3 independent and characterized by cytoplasmic vacuolation and no distinct nuclear fragmentation that is to be privileged in the treatment of apoptotic resistance pathways in triple-negative breast cancers (TNBCs).
Collapse
Affiliation(s)
- Malihe Behzadi
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335, Tehran, Iran
| | - Mehriar Amininasab
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, 16 Azar Street, 14174-66191, Tehran, Iran
| | - Marzieh Eghtedardoost
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-111, Tehran, Tehran, Iran
| | - Mojtaba Bagheri
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335, Tehran, Iran.
| |
Collapse
|
13
|
Nasiri F, Atanaki FF, Behrouzi S, Kavousi K, Bagheri M. CpACpP: In Silico Cell-Penetrating Anticancer Peptide Prediction Using a Novel Bioinformatics Framework. ACS OMEGA 2021; 6:19846-19859. [PMID: 34368571 PMCID: PMC8340416 DOI: 10.1021/acsomega.1c02569] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/13/2021] [Indexed: 05/12/2023]
Abstract
Cell-penetrating anticancer peptides (Cp-ACPs) are considered promising candidates in solid tumor and hematologic cancer therapies. Current approaches for the design and discovery of Cp-ACPs trust the expensive high-throughput screenings that often give rise to multiple obstacles, including instrumentation adaptation and experimental handling. The application of machine learning (ML) tools developed for peptide activity prediction is importantly of growing interest. In this study, we applied the random forest (RF)-, support vector machine (SVM)-, and eXtreme gradient boosting (XGBoost)-based algorithms to predict the active Cp-ACPs using an experimentally validated data set. The model, CpACpP, was developed on the basis of two independent cell-penetrating peptide (CPP) and anticancer peptide (ACP) subpredictors. Various compositional and physiochemical-based features were combined or selected using the multilayered recursive feature elimination (RFE) method for both data sets. Our results showed that the ACP subclassifiers obtain a mean performance accuracy (ACC) of 0.98 with an area under curve (AUC) ≈ 0.98 vis-à-vis the CPP predictors displaying relevant values of ∼0.94 and ∼0.95 via the hybrid-based features and independent data sets, respectively. Also, the predicting evaluation of Cp-ACPs gave accuracies of ∼0.79 and 0.89 on a series of independent sequences by applying our CPP and ACP classifiers, respectively, which leaves the performance of our predictors better than the earlier reported ACPred, mACPpred, MLCPP, and CPPred-RF. The described consensus-based fusion method additionally reached an AUC of 0.94 for the prediction of Cp-ACP (http://cbb1.ut.ac.ir/CpACpP/Index).
Collapse
Affiliation(s)
- Farid Nasiri
- Peptide
Chemistry Laboratory, Department of Biochemistry, Institute of Biochemistry
and Biophysics (IBB), University of Tehran, Tehran 14176-14335, Iran
| | - Fereshteh Fallah Atanaki
- Laboratory
of Complex Biological Systems and Bioinformatics (CBB), Department
of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 14176-14411, Iran
| | - Saman Behrouzi
- Laboratory
of Complex Biological Systems and Bioinformatics (CBB), Department
of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 14176-14411, Iran
| | - Kaveh Kavousi
- Laboratory
of Complex Biological Systems and Bioinformatics (CBB), Department
of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 14176-14411, Iran
| | - Mojtaba Bagheri
- Peptide
Chemistry Laboratory, Department of Biochemistry, Institute of Biochemistry
and Biophysics (IBB), University of Tehran, Tehran 14176-14335, Iran
| |
Collapse
|