1
|
Alaei E, Hashemi F, Farahani N, Tahmasebi S, Nabavi N, Daneshi S, Mahmoodieh B, Rahimzadeh P, Taheriazam A, Hashemi M. Peptides in breast cancer therapy: From mechanisms to emerging drug delivery and immunotherapy strategies. Pathol Res Pract 2025; 269:155946. [PMID: 40174279 DOI: 10.1016/j.prp.2025.155946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Breast cancer therapy can be improved by the application of multifunctional peptides and they have unique features, such as high specificity, minimized toxicity, and the capability to influence diverse processes. The role of peptides in breas cancer therapy is highlighted in the present review, examining their functions as therapeutic agents, diagnostic tools, and drug delivery application. Therapeutic peptides have displayed the ability to regulate key pathways in breast tumor, including HER2, VEGF, and EGFR, providing ideal alternatives to the conventional chemotherapy with reduced adverse effects. Additionally, peptide-based vaccines and immune-modulating peptides have demonstrated the capacity in enhancing anti-cancer immunity. The incorporation of peptides into nanoparticles has improved the delivery of drugs and genes, enhanced anti-cancer efficacy while minimizing side impacts. The progresses in the peptide engineering, including stapled peptides, peptide-drug conjugates, and cell-penetrating peptides, have remarkably increased their therapeutic efficacy and stability, elevating their applications in breast cancer therapy. Peptides can be developed using bioinformatics and high-throughput screening technologies to optimize pharmacokinetics and bioavailability. Despite their promise, peptides demonstrate challenges such as enzymatic degradation, limited stability, and high production costs. These obstacles can be addressed through strategies such as peptide cyclization, the employement of non-natural amino acids, and nanoparticle encapsulation. This review explores these recent advancements and strategies, providing ideal insights into the clinical potential of peptides in breast tumor therapy.
Collapse
Affiliation(s)
- Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Zhang Y, Zhang M, Leng L, Wu Y, Yang H, Wang L, Liu B, Yang S, Yang Z, Chen S, Song C. Karyotype and genome size analyses for two spiders of the lycosidae family. Front Genet 2025; 16:1544087. [PMID: 40201569 PMCID: PMC11975668 DOI: 10.3389/fgene.2025.1544087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Background Karyotype and genome size are critical genetic characteristics with significant value for cytogenetics, taxonomy, phylogenetics, evolution, and molecular biology. The Lycosidae family, known for its diverse spiders with varying ecological habits and behavioral traits, has seen limited exploration of its karyotype and genome size. Methods We utilized an improved tissue drop technique to prepare chromosome slides and compare the features of male and female karyotypes for two wolf spiders with different habits of Lycosidae. Furthermore, we predicted their genome sizes using flow cytometry (FCM) and K-mer analysis. Results The karyotypes of female and male Hippasa lycosina were 2n♀ = 26 = 14 m + 12 sm and 2n♂ = 24 = 10 m + 14 sm, respectively, and were composed of metacentric (m) and submetacentric (sm) chromosomes. In contrast, the karyotypes of Lycosa grahami consisted of telocentric (t) and subtelocentric (st) chromosomes (2n♀ = 20 = 20th and 2n♂ = 18 = 12th + 6t, for females and males). The sex chromosomes were both X1X2O. The estimated sizes of the H. lycosina and L. grahami genomes were 1966.54-2099.89 Mb and 3692.81-4012.56 Mb, respectively. Flow cytometry yielded slightly smaller estimates for genome size compared to k-mer analysis. K-mer analysis revealed a genome heterozygosity of 0.42% for H. lycosina and 0.80% for L. grahami, along with duplication ratios of 21.39% and 54.91%, respectively. Conclusion This study describes the first analysis of the genome sizes and karyotypes of two spiders from the Lycosidae that exhibit differential habits and provides essential data for future phylogenetic, cytogenetic, and genomic studies.
Collapse
Affiliation(s)
- Yuxuan Zhang
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Mengying Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, China
| | - Liang Leng
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Ya Wu
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Hanting Yang
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Liangting Wang
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Baimei Liu
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Shuai Yang
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Zizhong Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, China
| | - Shilin Chen
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| | - Chi Song
- School of Pharmacy, Chengdu university of Traditional Chinese Medicine, Chengdu, China
- Institute of Herbgenomics, Chengdu university of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Zou L, Hou Y, Nie X, Wang S, Tian S, Sun Z, Sun Z, Xu X, Li G, Ma G, Liu H. All-Small-Molecule Supramolecular Hydrogel Combining Self-Delivery and ROS-Responsive Release for Inhibiting Tumor Growth and Postoperative Recurrence. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39993162 DOI: 10.1021/acsami.4c20852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Supramolecular hydrogels show unprecedented advantages and have attracted widespread attention in biomedical sciences. However, it is challenging for bioactive star molecules, such as celastrol, to meet ideal formation conditions. Here, we report a dynamic covalent method to construct a dihydrol-type celastrol-phenylenediboronic acid-guanosine (DHcelPBG) supramolecular hydrogel. The DHcelPBG hydrogel can effectively accelerate 4T1 cell apoptosis by modulating the PI3K/Akt signaling pathway. Especially, the DHcelPBG hydrogel can serve as a self-delivery platform for reactive oxygen species (ROS)-facilitated self-release. An excessive ROS-containing tumor microenvironment can promote the obtained DHcelPBG hydrogel to kill more 4T1 tumor cells. Meanwhile, the hydrogel also exhibits distinguished degradability and biocompatibility. Subsequently, the orthotopic 4T1 tumor model results further demonstrate that the DHcelPBG hydrogel remarkably inhibits tumor growth and does not damage healthy tissue. In the postoperative recurrence 4T1 tumor model, the DHcelPBG hydrogel also effectively prevents postoperative tumor recurrence and lung metastasis without causing adverse side effects, resulting in an extended lifetime. The DHcelPBG hydrogel also exhibits distinguished degradability and biocompatibility. The DHcelPBG hydrogel integrates ROS-responsiveness, localized self-delivery, and antitumor activity into one system for breast cancer treatment with fewer side effects, showing great potential for clinical transformation in cancer therapy.
Collapse
Affiliation(s)
- Linjun Zou
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Yong Hou
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Xueqiang Nie
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Shengchen Wang
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Sichao Tian
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Zhaocui Sun
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Zhonghao Sun
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xudong Xu
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Guang Li
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan Branch, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Jinghong 666100, China
| | - Guoxu Ma
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Haitao Liu
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
4
|
dos Santos DM, Santiago LR, dos Santos NA, Romão W, Resende JM, de Lima ME, Borges MH, Ribeiro RIMDA. Encapsulated LyeTx III Peptide: Cytotoxic Agent Isolated from Lycosa erythrognatha Spider Venom. Toxins (Basel) 2025; 17:32. [PMID: 39852985 PMCID: PMC11768708 DOI: 10.3390/toxins17010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
The discovery of novel cytotoxic drugs is of paramount importance in contemporary medical research, particularly in the search for treatments with fewer side effects and higher specificity. Antimicrobial peptides are an interesting class of molecules for this endeavor. In this context, the LyeTx III, a new peptide extracted from the venom of the Lycosa erythrognatha spider, stands out. The peptide exhibits typical antimicrobial traits: a positive net charge and amphipathic α -helix structure in lipid-like environments. Its unique sequence (GKAMKAIAKFLGR-NH2), identified via mass spectrometry and Edman degradation, shows limited similarity to existing peptides. Significantly, when liposome-encapsulated, LyeTx III demonstrates selective activity against tumor cells in culture. Our MTT results showed that the cytotoxicity of the peptide increased against HN13 cells when administered as liposomes, with their viability in HN13 cells alone being 98%, compared to 38% in liposome-encapsulated form. This finding underscores that the LyeTx III peptide may be a good candidate for the development of new drugs against cancer. Its activity when encapsulated is promising, as it can increase its half-life in the body and can also be targeted to specific tumors.
Collapse
Affiliation(s)
- Daniel Moreira dos Santos
- Department of Experimental Pathology, Federal University of São João del-Rei, Divinópolis 36301-158, Brazil; (D.M.d.S.); (L.R.S.)
| | - Livia Ramos Santiago
- Department of Experimental Pathology, Federal University of São João del-Rei, Divinópolis 36301-158, Brazil; (D.M.d.S.); (L.R.S.)
| | - Nayara Araújo dos Santos
- Petroleomics and Forensics Laboratory, Federal University of Espírito Santo, Vitória 29075-910, Brazil; (N.A.d.S.); (W.R.)
| | - Wanderson Romão
- Petroleomics and Forensics Laboratory, Federal University of Espírito Santo, Vitória 29075-910, Brazil; (N.A.d.S.); (W.R.)
| | - Jarbas Magalhães Resende
- Department of Chemistry, Federal University of Minas Gerais (UFMG), Belo Horizonte 30110-005, Brazil;
| | - Maria Elena de Lima
- Programa de Pós-Graduação em Medicina-Biomedicina, Faculdade Santa Casa de Belo Horizonte, Belo Horizonte 30110-005, Brazil
| | - Márcia Helena Borges
- Proteomics and Arachnid Laboratory, Ezequiel Dias Foundation, Belo Horizonte 30110-005, Brazil;
| | | |
Collapse
|
5
|
Souza JLN, Antunes-Porto AR, da Silva Oliveira I, Amorim CCO, Pires LO, de Brito Duval I, Amaral LVBD, Souza FR, Oliveira EA, Cassali GD, Cardoso VN, Fernandes SOA, Fujiwara RT, Russo RC, Bueno LL. Screening and validating the optimal panel of housekeeping genes for 4T1 breast carcinoma and metastasis studies in mice. Sci Rep 2024; 14:26476. [PMID: 39488625 PMCID: PMC11531515 DOI: 10.1038/s41598-024-77126-x] [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: 05/17/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
The 4T1 model is extensively employed in murine studies to elucidate the mechanisms underlying the carcinogenesis of triple-negative breast cancer. Molecular biology serves as a cornerstone in these investigations. However, accurate gene expression analyses necessitate data normalization employing housekeeping genes (HKGs) to avert spurious results. Here, we initially delve into the characteristics of the tumor evolution induced by 4T1 in mice, underscoring the imperative for additional tools for tumor monitoring and assessment methods for tracking the animals, thereby facilitating prospective studies employing this methodology. Subsequently, leveraging various software platforms, we assessed ten distinct HKGs (GAPDH, 18 S, ACTB, HPRT1, B2M, GUSB, PGK1, CCSER2, SYMPK, ANKRD17) not hitherto evaluated in the 4T1 breast cancer model, across tumors and diverse tissues afflicted by metastasis. Our principal findings underscore GAPDH as the optimal HKG for gene expression analyses in tumors, while HPRT1 emerged as the most stable in the liver and CCSER2 in the lung. These genes demonstrated consistent expression and minimal variation among experimental groups. Furthermore, employing these HKGs for normalization, we assessed TNF-α and VEGF expression in tissues and discerned significant disparities among groups. We posit that this constitutes the inaugural delineation of an ideal HKG for experiments utilizing the 4T1 model, particularly in vivo settings.
Collapse
Affiliation(s)
- Jorge Lucas Nascimento Souza
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Rafaela Antunes-Porto
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela da Silva Oliveira
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Chiara Cássia Oliveira Amorim
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Octávio Pires
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela de Brito Duval
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luisa Vitor Braga do Amaral
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Rezende Souza
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Evelyn Ane Oliveira
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Valbert Nascimento Cardoso
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone Odília Antunes Fernandes
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270- 901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
6
|
Freuville L, Matthys C, Quinton L, Gillet JP. Venom-derived peptides for breaking through the glass ceiling of drug development. Front Chem 2024; 12:1465459. [PMID: 39398192 PMCID: PMC11468230 DOI: 10.3389/fchem.2024.1465459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Venoms are complex mixtures produced by animals and consist of hundreds of components including small molecules, peptides, and enzymes selected for effectiveness and efficacy over millions of years of evolution. With the development of venomics, which combines genomics, transcriptomics, and proteomics to study animal venoms and their effects deeply, researchers have identified molecules that selectively and effectively act against membrane targets, such as ion channels and G protein-coupled receptors. Due to their remarkable physico-chemical properties, these molecules represent a credible source of new lead compounds. Today, not less than 11 approved venom-derived drugs are on the market. In this review, we aimed to highlight the advances in the use of venom peptides in the treatment of diseases such as neurological disorders, cardiovascular diseases, or cancer. We report on the origin and activity of the peptides already approved and provide a comprehensive overview of those still in development.
Collapse
Affiliation(s)
- Lou Freuville
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Chloé Matthys
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
7
|
Bashi M, Madanchi H, Yousefi B. Investigation of cytotoxic effect and action mechanism of a synthetic peptide derivative of rabbit cathelicidin against MDA-MB-231 breast cancer cell line. Sci Rep 2024; 14:13497. [PMID: 38866982 PMCID: PMC11169400 DOI: 10.1038/s41598-024-64400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024] Open
Abstract
Antimicrobial peptides (AMPs) have sparked significant interest as potential anti-cancer agents, thereby becoming a focal point in pursuing novel cancer-fighting strategies. These peptides possess distinctive properties, underscoring the importance of developing more potent and selectively targeted versions with diverse mechanisms of action against human cancer cells. Such advancements would offer notable advantages compared to existing cancer therapies. This research aimed to examine the toxicity and selectivity of the nrCap18 peptide in both cancer and normal cell lines. Furthermore, the rate of cellular death was assessed using apoptosis and acridine orange/ethidium bromide (AO/EB) double staining at three distinct incubation times. Additionally, the impact of this peptide on the cancer cell cycle and migration was evaluated, and ultimately, the expression of cyclin-dependent kinase 4/6 (CDK4/6) genes was investigated. The results obtained from the study demonstrated significant toxicity and selectivity in cancer cells compared to normal cells. Moreover, a strong progressive increase in cell death was observed over time. Furthermore, the peptide exhibited the ability to halt the progression of cancer cells in the G1 phase of the cell cycle and impede their migration by suppressing the expression of CDK4/6 genes.
Collapse
Affiliation(s)
- Marzieh Bashi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamid Madanchi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Biotechnology, School of Medicine, Semnan University of Medical Sciences, Semnan, 35131-38111, Iran.
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 13198, Iran.
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran.
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
8
|
Vitarelli da Silva T, Bernardes D, Oliveira-Lima OC, Fernandes Pinto B, Limborço Filho M, Fraga Faraco CC, Juliano MA, Esteves Arantes RM, A Moreira F, Carvalho-Tavares J. Cannabidiol Attenuates In Vivo Leukocyte Recruitment to the Spinal Cord Microvasculature at Peak Disease of Experimental Autoimmune Encephalomyelitis. Cannabis Cannabinoid Res 2024; 9:537-546. [PMID: 36745386 DOI: 10.1089/can.2022.0103] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Introduction: Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system characterized by neuroinflammation leading to demyelination. The associated symptoms lead to a devastating decrease in quality of life. The cannabinoids and their derivatives have emerged as an encouraging alternative due to their management of symptom in MS. Objective: The aim of the study was to investigate the mechanism of action of cannabidiol (CBD), a nonpsychoactive cannabinoid, on molecular and cellular events associated with leukocyte recruitment induced by experimental autoimmune encephalomyelitis (EAE). Materials and Methods: C57BL/6 female mice were randomly assigned to the four experimental groups: C (control group), CBD (cannabidiol-treated group, 5 mg/kg i.p.; 14 days), EAE (experimental autoimmune encephalomyelitis-induced group), and EAE+CBD (experimental autoimmune encephalomyelitis-induced plus cannabidiol-treated group). Results: The results indicated that 5 mg/kg of CBD injected intraperitoneally between the 1st and 14th days of EAE could reduce the leukocyte rolling and adhesion into the spinal cord microvasculature as well cellular tissue infiltration. These results were supported by a decreased mRNA expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in the spinal cord. Conclusion: Purified CBD reduces in vivo VCAM and ICAM-mediated leukocyte recruitment to the spinal cord microvasculature at EAE peak disease.
Collapse
Affiliation(s)
- Thiago Vitarelli da Silva
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danielle Bernardes
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade de Campinas, Campinas, Brazil
| | - Onésia Cristina Oliveira-Lima
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratório de Neuroquímica e Neurofarmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Bárbara Fernandes Pinto
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Limborço Filho
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Cristina Fraga Faraco
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Aparecida Juliano
- Enzimas proteolíticas e Síntese de peptídeos, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rosa Maria Esteves Arantes
- Neuroimunopatologia Experimental, Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício A Moreira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Carvalho-Tavares
- Núcleo de Neurociências, Programa de Pós-graduação em Ciências Biológicas:Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Núcleo de Educação e Comunicação em Ciências da Vida e da Saúde (NEDUCOM), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Díaz-Gómez JL, Martín-Estal I, Rivera-Aboytes E, Gaxiola-Muñíz RA, Puente-Garza CA, García-Lara S, Castorena-Torres F. Biomedical applications of synthetic peptides derived from venom of animal origin: A systematic review. Biomed Pharmacother 2024; 170:116015. [PMID: 38113629 DOI: 10.1016/j.biopha.2023.116015] [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: 10/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Development of therapeutic agents that have fewer adverse effects and have higher efficacy for diseases, such as cancer, metabolic disorders, neurological diseases, infections, cardiovascular diseases, and respiratory diseases, are required. Recent studies have focused on identifying novel sources for pharmaceutical molecules to develop therapies against these diseases. Among the sources for potentially new therapies, animal venom-derived molecules have generated much interest. Various animal venom-derived proteins and peptides have been isolated, identified, synthesized, and tested to develop drugs. Venom-derived peptides have several biomedical properties, such as proapoptotic, cell migration, and autophagy regulation activities in cancer cell models; induction of vasodilation by nitric oxide and regulation of angiotensin II; modification of insulin response by controlling calcium and potassium channels; regulation of pain receptor activity; modulation of immune cell activity; alteration of motor neuron activity; degradation or inhibition of β-amyloid plaque formation; antibacterial, antifungal, antiviral, and antiprotozoal activities; increase in sperm motility and potentiation of erectile function; reduction of intraocular pressure; anticoagulation, fibrinolytic, and antithrombotic activities; etc. This systematic review compiles these biomedical properties and potential biomedical applications of synthesized animal venom-derived peptides reported in the latest research. In addition, the limitations and areas of opportunity in this research field are discussed so that new studies can be developed based on the data presented.
Collapse
Affiliation(s)
- Jorge L Díaz-Gómez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Elizabeth Rivera-Aboytes
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Ramón Alonso Gaxiola-Muñíz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - César A Puente-Garza
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Silverio García-Lara
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Fabiola Castorena-Torres
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico.
| |
Collapse
|
10
|
Mohammadi N, Nouri F, Asgari Y, Moradi-Sardareh H, Sharafi-Kolkeshvandi M, Nemati H, Kardar GA. The immunostimulant effects of the rice ragged stunt virus genome on the growth and metastasis of breast cancer in mouse model. Int Immunopharmacol 2023; 125:111101. [PMID: 37922568 DOI: 10.1016/j.intimp.2023.111101] [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: 06/08/2023] [Revised: 09/30/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023]
Abstract
There are multiple treatment strategies that have been reported for breast cancer, while new and effective therapies against it are still necessary. Stimulating the immune system and its components against cancer cells is one of the unique treatment strategies of immunotherapy and long dsRNAs are immunostimulant in this regard. Based on bioinformatics approaches, a fragment of the Rice ragged stunt RNA virus genome was selected and synthesized according to its immunogenicity. Based on the in vitro transcription technique, dsRNA was synthesized and its binding ability to the PEI/PEI-Ac Polyethylenimine (PEI) or Acetylated polyethylenimine (PEI-Ac) was verified by the gel retardation assay. Then, the PEI-Ac was synthesized by adding acetyl groups to the PEI, and the results of the 1H NMR method indicated its successful synthesis. After cancer induction by 4 T1 cells in Balb/C mice, intraperitoneal (IP) and intratumoral (IT) treatment by the PEI/PEI-Ac-dsRNA were performed and the tumor growth inhibition was evaluated. Results demonstrated that PEI/PEI-Ac-dsRNA can lead to a decrease in tumor weight and volume in both the IP and IT routes. Also, by using macro-metastatic nodule counting and hematoxylin and eosin (H&E) staining we showed that PEI/PEI-Ac-dsRNA can prevent micro and macro-metastasis in the lung. Therefore, the PEI/PEI-Ac-dsRNA acts as an effective inhibitor of growth and metastasis of the breast cancer models. We showed that viral dsRNA can exert its antitumor properties by stimulating TNF-α and IFN-γ. In general, our results revealed that dsRNA derived from the plant virus genome stimulates the intrinsic immune system and can be a potential immune stimulant drug for cancer treatment.
Collapse
Affiliation(s)
- Nejad Mohammadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Immunology Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Nouri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yazdan Asgari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hemen Moradi-Sardareh
- Asadabad School of Medicine, Hamadan University of Medical Sciences, Asadabad, Iran; BioMad AS Company, Oslo, Norway
| | | | - Hossein Nemati
- Genetic Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Gholam Ali Kardar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Immunology Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Chen PJ, Lin ES, Su HH, Huang CY. Cytotoxic, Antibacterial, and Antioxidant Activities of the Leaf Extract of Sinningia bullata. PLANTS (BASEL, SWITZERLAND) 2023; 12:859. [PMID: 36840206 PMCID: PMC9967939 DOI: 10.3390/plants12040859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 06/12/2023]
Abstract
Sinningia bullata is a tuberous member of the flowering plant family Gesneriaceae. Prior to this work, the antibacterial, antioxidant, and cytotoxic properties of S. bullata were undetermined. Here, we prepared different extracts from the leaf, stem, and tuber of S. bullata and investigated their pharmacological activities. The leaf extract of S. bullata, obtained by 100% acetone (Sb-L-A), had the highest total flavonoid content, antioxidation capacity, and cytotoxic and antibacterial activities. Sb-L-A displayed a broad range of antibacterial activities against Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa. The inhibition zones of Sb-L-A ranged from 8 to 30 mm and were in the following order: S. aureus > E. coli > P. aeruginosa. Incubation of B16F10 melanoma cells with Sb-L-A at a concentration of 80 μg/mL caused deaths at the rate of 96%, reduced migration by 100%, suppressed proliferation and colony formation by 99%, and induced apoptosis, which was observed in 96% of the B16F10 cells. In addition, the cytotoxic activities of Sb-L-A were synergistically enhanced when coacting with the antitumor drug epothilone B. Sb-L-A was also used to determine the cytotoxic effects against 4T1 mammary carcinoma cells. Sb-L-A of 60 μg/mL boosted the distribution of the G2 phase from 1.4% to 24.4% in the B16F10 cells. Accordingly, Sb-L-A might suppress melanoma cell proliferation by inducing G2 cell-cycle arrest. The most abundant compounds in Sb-L-A were identified using gas chromatography-mass spectrometry. Overall, the collective data in this study may indicate the pharmacological potentials of Sb-L-A for possible medical applications.
Collapse
Affiliation(s)
- Pin-Jui Chen
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan;
| | - En-Shyh Lin
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung City 403, Taiwan;
| | - Hsin-Hui Su
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 717, Taiwan;
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
12
|
Huang YH, Chiang WY, Chen PJ, Lin ES, Huang CY. Anticancer and Antioxidant Activities of the Root Extract of the Carnivorous Pitcher Plant Sarracenia purpurea. PLANTS (BASEL, SWITZERLAND) 2022; 11:1668. [PMID: 35807620 PMCID: PMC9269354 DOI: 10.3390/plants11131668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 05/27/2023]
Abstract
The carnivorous pitcher plant Sarracenia purpurea exhibits many ethnobotanical uses, including the treatments of type 2 diabetes and tuberculosis-like symptoms. In this study, we prepared different extracts from the leaves (pitchers), stems, and roots of S. purpurea and investigated their antioxidant and anticancer properties. To evaluate the extraction efficiency, we individually used different solvents, namely methanol, ethanol, acetone, and distilled water, for S. purpurea extract preparations. The root extract of S. purpurea, obtained by 100% acetone (S. purpurea-root-acetone), had the highest anticancer activities, antioxidation capacity (the DPPH activity with IC50 of 89.3 ± 2.2 μg/mL), antibacterial activities, total phenolic content (33.4 ± 0.7 mg GAE/g), and total flavonoid content (107.9 ± 2.2 mg QUE/g). The most abundant compounds in S. purpurea-root-acetone were identified using gas chromatography-mass spectrometry; 7,8-Dihydro-α-ionone was the major compound present in S. purpurea-root-acetone. In addition, the co-cytotoxicity of S. purpurea-root-acetone (combined with the clinical anticancer drug 5-fluorouracil (5-FU) on the survival, apoptosis, proliferation, and migration of the 4T1 mammary carcinoma) was examined. The combination of 5-FU with S. purpurea-root-acetone could be highly efficient for anti-4T1 cells. We also found that S. purpurea-root-acetone could inhibit the enzymatic activity of human dihydroorotase (huDHOase), an attractive target for potential anticancer chemotherapy. The sic most abundant compounds in S. purpurea-root-acetone were tested using an in silico analysis via MOE-Dock software for their binding affinities. The top-ranked docking conformations were observed for 7,8-dihydro-α-ionone and stigmast-5-en-3-ol, suggesting the inhibition potential against huDHOase. Overall, the collective data in this study may indicate the pharmacological potentials of S. purpurea-root-acetone for possible medical applications.
Collapse
Affiliation(s)
- Yen-Hua Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan; (Y.-H.H.); (W.-Y.C.); (P.-J.C.)
| | - Wei-Yu Chiang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan; (Y.-H.H.); (W.-Y.C.); (P.-J.C.)
| | - Pin-Jui Chen
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan; (Y.-H.H.); (W.-Y.C.); (P.-J.C.)
| | - En-Shyh Lin
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung City 403, Taiwan;
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan; (Y.-H.H.); (W.-Y.C.); (P.-J.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
13
|
de Avelar Júnior JT, Lima-Batista E, Castro Junior CJ, Pimenta AMDC, Dos Santos RG, Souza-Fagundes EM, De Lima ME. LyeTxI-b, a Synthetic Peptide Derived From a Spider Venom, Is Highly Active in Triple-Negative Breast Cancer Cells and Acts Synergistically With Cisplatin. Front Mol Biosci 2022; 9:876833. [PMID: 35601827 PMCID: PMC9114809 DOI: 10.3389/fmolb.2022.876833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/04/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common cancer that affects women globally and is among the leading cause of women’s death. Triple-negative breast cancer is more difficult to treat because hormone therapy is not available for this subset of cancer. The well-established therapy against triple-negative breast cancer is mainly based on surgery, chemotherapy, and immunotherapy. Among the drugs used in the therapy are cisplatin and carboplatin. However, they cause severe toxicity to the kidneys and brain and cause nausea. Therefore, it is urgent to propose new chemotherapy techniques that provide new treatment options to patients affected by this disease. Nowadays, peptide drugs are emerging as a class of promising new anticancer agents due to their lytic nature and, apparently, a minor drug resistance compared to other conventional drugs (reviewed in Jafari et al., 2022). We have recently reported the cytotoxic effect of the antimicrobial peptide LyeTx I-b against glioblastoma cells (Abdel-Salam et al., 2019). In this research, we demonstrated the cytotoxic effect of the peptide LyeTx I-b, alone and combined with cisplatin, against triple-negative cell lines (MDA-MD-231). LyeTx-I-b showed a selectivity index 70-fold higher than cisplatin. The peptide:cisplatin combination (P:C) 1:1 presented a synergistic effect on the cell death and a selective index value 16 times greater than the cisplatin alone treatment. Therefore, an equi-effective reduction of cisplatin can be reached in the presence of LyeTx I-b. Cells treated with P:C combinations were arrested in the G2/M cell cycle phase and showed positive staining for acridine orange, which was inhibited by bafilomycin A1, indicating autophagic cell death (ACD) as a probable cell death mechanism. Furthermore, Western blot experiments indicated a decrease in P21 expression and AKT phosphorylation. The decrease in AKT phosphorylation is indicative of ACD. However, other studies are still necessary to better elucidate the pathways involved in the cell death mechanism induced by the peptide and the drug combinations. These findings confirmed that the peptide LyeTx I-b seems to be a good candidate for combined chemotherapy to treat breast cancer. In addition, in vivo studies are essential to validate the use of LyeTx I-b as a therapeutic drug candidate, alone and/or combined with cisplatin.
Collapse
Affiliation(s)
- Joaquim Teixeira de Avelar Júnior
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Joaquim Teixeira de Avelar Júnior, ; Maria Elena De Lima,
| | - Edleusa Lima-Batista
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Célio José Castro Junior
- Programa de Pós-Graduação em Medicina e Biomedicina da Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
| | | | | | - Elaine Maria Souza-Fagundes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Elena De Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Programa de Pós-Graduação em Medicina e Biomedicina da Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
- *Correspondence: Joaquim Teixeira de Avelar Júnior, ; Maria Elena De Lima,
| |
Collapse
|