1
|
Bhardwaj P, Raigond B, Raigond P, Verma A, Verma G, Kochhar T, Patroti P, Das IK, Satyavathi CT. Antiviral activity of ribosome inactivating proteins for management of plant viral infection. Virology 2025; 603:110403. [PMID: 39894605 DOI: 10.1016/j.virol.2025.110403] [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: 09/19/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025]
Abstract
In nature, plants exhibit various defense mechanisms to protect themselves from viral infection. Reported to harbor virus-inhibiting compounds like Ribosome inactivating proteins (RIPs). It's a matter of how we explore, identify, and utilize RIPs in managing a given stress. RIPs have been found to contain antiviral, anticancer, and neurotoxic effects and are used in various biomedical and agricultural fields. The expression of RIPs could be enhanced in plants to improve their defense against biotic and abiotic stresses. Identification of new RIPs and genetic sequencing led to the development of new phylogenetic theories. Studies on the interaction between RIPs and cells have increased the knowledge regarding the handling of exogenous proteins by cells. The review provides a brief historical preview, classification, mode of action, and broader applications with a special focus on managing plant viral diseases and concerns to mankind.
Collapse
Affiliation(s)
- Pooja Bhardwaj
- ICAR-Central Potato Research Institute, Shimla, 171001, Himachal Pradesh, India
| | - Baswaraj Raigond
- ICAR-Central Potato Research Institute, Shimla, 171001, Himachal Pradesh, India; Centre for Rabi Sorghum, ICAR-Indian Institute of Millets Research, Regional Station, Solapur, 413006, Maharashtra, India.
| | - Pinky Raigond
- ICAR-Central Potato Research Institute, Shimla, 171001, Himachal Pradesh, India; ICAR-National Research Centre on Pomegranate, Solapur, 413255, Maharashtra, India
| | - Ambika Verma
- ICAR-Central Potato Research Institute, Shimla, 171001, Himachal Pradesh, India
| | - Gaurav Verma
- ICAR-Vivekananda Parvatiya Krishi Anusandhan Sansthan, Almora, 263601, Uttarakhand, India
| | - Tarvinder Kochhar
- ICAR-Central Potato Research Institute, Shimla, 171001, Himachal Pradesh, India
| | - Parashuram Patroti
- Centre for Rabi Sorghum, ICAR-Indian Institute of Millets Research, Regional Station, Solapur, 413006, Maharashtra, India
| | - I K Das
- ICAR- ICAR-Indian Institute of Millets Research, Hyderabad, 500030, Telangana, India
| | - C Tara Satyavathi
- ICAR- ICAR-Indian Institute of Millets Research, Hyderabad, 500030, Telangana, India
| |
Collapse
|
2
|
Shi Y, Shi M, Wang Y, You J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther 2024; 9:322. [PMID: 39543114 PMCID: PMC11564800 DOI: 10.1038/s41392-024-02002-z] [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: 06/02/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
In the last decade, messenger ribonucleic acid (mRNA)-based drugs have gained great interest in both immunotherapy and non-immunogenic applications. This surge in interest can be largely attributed to the demonstration of distinct advantages offered by various mRNA molecules, alongside the rapid advancements in nucleic acid delivery systems. It is noteworthy that the immunogenicity of mRNA drugs presents a double-edged sword. In the context of immunotherapy, extra supplementation of adjuvant is generally required for induction of robust immune responses. Conversely, in non-immunotherapeutic scenarios, immune activation is unwanted considering the host tolerability and high expression demand for mRNA-encoded functional proteins. Herein, mainly focused on the linear non-replicating mRNA, we overview the preclinical and clinical progress and prospects of mRNA medicines encompassing vaccines and other therapeutics. We also highlight the importance of focusing on the host-specific variations, including age, gender, pathological condition, and concurrent medication of individual patient, for maximized efficacy and safety upon mRNA administration. Furthermore, we deliberate on the potential challenges that mRNA drugs may encounter in the realm of disease treatment, the current endeavors of improvement, as well as the application prospects for future advancements. Overall, this review aims to present a comprehensive understanding of mRNA-based therapies while illuminating the prospective development and clinical application of mRNA drugs.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Meixing Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, P. R. China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, P. R. China.
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, P. R. China.
| |
Collapse
|
3
|
Faraz A, Amani J, Arbabian S, Karizi SZ, Torbati MB. In vitro analysis of single chain variable fragment-based immunotoxins against Erythropoietin-producing hepatocellular A2 receptor overexpressed in breast cancer cells. J Immunol Methods 2024; 533:113732. [PMID: 39116931 DOI: 10.1016/j.jim.2024.113732] [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: 11/29/2023] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Breast cancer is one of the leading causes of cancer deaths worldwide. Thereafter, designing new treatments with higher specificity and efficacy is urgently required. In this regard, targeted immunotherapy using immunotoxins has shown great promise in treating cancer. To target a breast cancer cell, the authors used the antibody fragment against a receptor tyrosine kinase, EphA2, which is overexpressed in many cancers. This fragment was conjugated to a plant toxin, subunit A of ricin, in two different orientations from N to C-terminal (EphA2- C-Ricin and EphA2- N-Ricin). Then, these two immunotoxins were characterized using in vitro cell-based assays. Three different cell lines were treated, MDA-MB-231 (breast cancer) which has a high level of EphA2 expression, MCF-7 (breast cancer) which has a low level of EphA2 expression, and HEK293 (human embryonic kidney) which has a very low level of EphA2 expression. Moreover, binding ability, cytotoxicity, internalization, and apoptosis capacity of these two newly developed immunotoxins were investigated. The flow cytometry using Annexin V- Propidium iodide (PI) method indicated significant induction of apoptosis only in the MDA-MB-231 cells at different concentrations. It was also found that construct I, EphA2- C-Ricin immunotoxin, could bind, internalize, and induce apoptosis better than the EphA2- N-Ricin immunotoxin. In addition, the obtained data suggested that the N or C-terminal orientation conformation is of significant importance.
Collapse
Affiliation(s)
- Atefeh Faraz
- Department of Cellular and Molecular Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Sedigheh Arbabian
- Department of Cellular and Molecular Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Shohreh Zare Karizi
- Department of Genetic and Biotechnology, Varamin Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Maryam Bikhof Torbati
- Department of Biology, Yadegar-e-Imam Khomeini (RAH) Shahr-e-Rey Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Oghalaie A, Hosseini ME, Hosseininejad-Chafi M, Eftekhari Z, Behdani M, Kazemi-Lomedasht F. Advances in immunotoxin engineering: precision therapeutic strategies in modern oncology. Med Oncol 2024; 41:239. [PMID: 39230639 DOI: 10.1007/s12032-024-02478-3] [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] [Accepted: 08/12/2024] [Indexed: 09/05/2024]
Abstract
Immunotoxins (ITs) are specialized therapeutic agents designed for targeted treatment, particularly in cancer therapy. They consist of a monoclonal antibody or antibody fragment linked to a potent cytotoxic agent, such as bacterial- or plant-derived toxins like diphtheria toxin, ricin, or pseudomonas exotoxin. The monoclonal antibody component specifically binds to antigens expressed on the surface of target cells, facilitating the internalization of the IT. Once inside the cell, the cytotoxic agent is released, disrupting essential cellular processes and leading to cell death. This targeted approach minimizes damage to healthy tissues while effectively eliminating diseased cells. The production of ITs involves two primary methods: recombinant fusion and chemical conjugation. In recombinant fusion, genetic engineering is used to create a fusion protein that combines the antibody and toxin, ensuring precise control over their ratio and functionality. In chemical conjugation, pre-existing antibodies are chemically linked to toxins, allowing for greater flexibility in combining different antibodies and cytotoxic agents. Each method has its advantages and challenges, influencing the specificity, production complexity, and therapeutic potential of the resulting ITs. As research advances, ITs continue to show promise not only in oncology but also in treating other diseases, including inflammatory conditions and atherosclerosis. The precise targeting and potent effects of ITs make them a valuable tool in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahmoud Eshagh Hosseini
- Gastroenterology and Liver Department, Amiralam Hospital, University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Wani SS, Qadri H, Shah AH, Dar TA. Dual Antifungal and Antiproliferative Activities of a Novel Protein Fraction from a Medicinally Important Herb Trillium govanianum Wall. ex. D. Don. Appl Biochem Biotechnol 2024; 196:5080-5098. [PMID: 38038807 DOI: 10.1007/s12010-023-04786-2] [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] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Antimicrobial resistance of microorganisms and the unwanted side effects of chemoradiation therapy in cancer are major issues in healthcare. In recent times, protein-based drugs have emerged as promising candidates due to their high specificity, less side effects, etc. In this context, the rhizome of Trillium govanianum was first explored for biologically active proteins/peptides. For this, three protein fractions namely Aqueous protein fraction (APF), Hexane-Methanol-treated aqueous protein fraction (HMAPF), and Methanol-treated aqueous protein fraction (MAPF) were prepared and evaluated for antimicrobial and antiproliferative activities. In antifungal activity, HMAPF showed the lowest MIC90 values of 1.56 µg/ml against Candida parapsilosis and Candida glabrata and 3.12 µg/ml against Candida albicans and Candida auris. The antifungal activity was further confirmed by a chitinase assay, a growth kinetics and a proteinase inhibitory assay. Surprisingly, none of the three protein fractions exhibited antibacterial activity against Escherichia coli and Staphylococcus aureus. Moreover, APF exhibited potent antiproliferative and antioxidant activities with IC50 values of 18 µg/ml and 227 µg /ml, respectively. For HMAPF, an IC50 value of 70 µg/ml against the MDA-MB-231 cell line was observed. The present results demonstrate that the protein fractions, particularly HMAPF and APF, might serve as potential sources of a dual antifungal and antiproliferative protein-based drug.
Collapse
Affiliation(s)
- Snober S Wani
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, India, 190006
| | - Hafsa Qadri
- Department of Bioresources, University of Kashmir, Srinagar, Jammu and Kashmir, India, 190006
| | - Abdul H Shah
- Department of Bioresources, University of Kashmir, Srinagar, Jammu and Kashmir, India, 190006.
| | - Tanveer A Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, India, 190006.
| |
Collapse
|
6
|
Schlaak L, Weise C, Kuropka B, Weng A. Mutational Analysis of RIP Type I Dianthin-30 Suggests a Role for Arg24 in Endocytosis. Toxins (Basel) 2024; 16:219. [PMID: 38787071 PMCID: PMC11125672 DOI: 10.3390/toxins16050219] [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: 01/26/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Saponin-mediated endosomal escape is a mechanism that increases the cytotoxicity of type I ribosome-inactivating proteins (type I RIPs). In order to actualize their cytotoxicity, type I RIPs must be released into the cytosol after endocytosis. Without release from the endosomes, type I RIPs are largely degraded and cannot exert their cytotoxic effects. Certain triterpene saponins are able to induce the endosomal escape of these type I RIPs, thus increasing their cytotoxicity. However, the molecular mechanism underlying the endosomal escape enhancement of type I RIPs by triterpene saponins has not been fully elucidated. In this report, we investigate the involvement of the basic amino acid residues of dianthin-30, a type I RIP isolated from the plant Dianthus caryophyllus L., in endosomal escape enhancement using alanine scanning. Therefore, we designed 19 alanine mutants of dianthin-30. Each mutant was combined with SO1861, a triterpene saponin isolated from the roots of Saponaria officinalis L., and subjected to a cytotoxicity screening in Neuro-2A cells. Cytotoxic screening revealed that dianthin-30 mutants with lysine substitutions did not impair the endosomal escape enhancement. There was one particular mutant dianthin, Arg24Ala, that exhibited significantly reduced synergistic cytotoxicity in three mammalian cell lines. However, this reduction was not based on an altered interaction with SO1861. It was, rather, due to the impaired endocytosis of dianthin Arg24Ala into the cells.
Collapse
Affiliation(s)
- Louisa Schlaak
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany;
| | - Christoph Weise
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany; (C.W.); (B.K.)
| | - Benno Kuropka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany; (C.W.); (B.K.)
| | - Alexander Weng
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany;
| |
Collapse
|
7
|
Iglesias R, Citores L, Gay CC, Ferreras JM. Antifungal Activity of Ribosome-Inactivating Proteins. Toxins (Basel) 2024; 16:192. [PMID: 38668617 PMCID: PMC11054410 DOI: 10.3390/toxins16040192] [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: 03/14/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
The control of crop diseases caused by fungi remains a major problem and there is a need to find effective fungicides that are environmentally friendly. Plants are an excellent source for this purpose because they have developed defense mechanisms to cope with fungal infections. Among the plant proteins that play a role in defense are ribosome-inactivating proteins (RIPs), enzymes obtained mainly from angiosperms that, in addition to inactivating ribosomes, have been studied as antiviral, fungicidal, and insecticidal proteins. In this review, we summarize and discuss the potential use of RIPs (and other proteins with similar activity) as antifungal agents, with special emphasis on RIP/fungus specificity, possible mechanisms of antifungal action, and the use of RIP genes to obtain fungus-resistant transgenic plants. It also highlights the fact that these proteins also have antiviral and insecticidal activity, which makes them very versatile tools for crop protection.
Collapse
Affiliation(s)
- Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (L.C.)
| | - Lucía Citores
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (L.C.)
| | - Claudia C. Gay
- Laboratory of Protein Research, Institute of Basic and Applied Chemistry of Northeast Argentina (UNNE-CONICET), Faculty of Exact and Natural Sciences and Surveying, Av. Libertad 5470, Corrientes 3400, Argentina;
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (L.C.)
| |
Collapse
|
8
|
di Leandro L, Colasante M, Pitari G, Ippoliti R. Hosts and Heterologous Expression Strategies of Recombinant Toxins for Therapeutic Purposes. Toxins (Basel) 2023; 15:699. [PMID: 38133203 PMCID: PMC10748335 DOI: 10.3390/toxins15120699] [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: 11/15/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The production of therapeutic recombinant toxins requires careful host cell selection. Bacteria, yeast, and mammalian cells are common choices, but no universal solution exists. Achieving the delicate balance in toxin production is crucial due to potential self-intoxication. Recombinant toxins from various sources find applications in antimicrobials, biotechnology, cancer drugs, and vaccines. "Toxin-based therapy" targets diseased cells using three strategies. Targeted cancer therapy, like antibody-toxin conjugates, fusion toxins, or "suicide gene therapy", can selectively eliminate cancer cells, leaving healthy cells unharmed. Notable toxins from various biological sources may be used as full-length toxins, as plant (saporin) or animal (melittin) toxins, or as isolated domains that are typical of bacterial toxins, including Pseudomonas Exotoxin A (PE) and diphtheria toxin (DT). This paper outlines toxin expression methods and system advantages and disadvantages, emphasizing host cell selection's critical role.
Collapse
Affiliation(s)
| | | | | | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.d.L.); (M.C.); (G.P.)
| |
Collapse
|
9
|
Sheikhalipour M, Gohari G, Esmaielpour B, Behnamian M, Giglou MT, Milani MH, Bahrami MK, Kulak M, Ioannou A, Fotopoulos V, Vita F. Effect of melatonin foliar sprays on morphophysiological attributes, fruit yield and quality of Momordica charantia L. under salinity stress. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 205:108194. [PMID: 37992418 DOI: 10.1016/j.plaphy.2023.108194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/20/2023] [Accepted: 11/12/2023] [Indexed: 11/24/2023]
Abstract
Soil salinity is one of the increasing problems in agricultural fields in many parts of the world, adversely affecting the performance and health of the plants. As a pleiotropic signal and antioxidant molecule in both animals and plants, melatonin has been reported to possess significant roles in combating with stress factors, in general and salt stress, in particular. In this study, the interactive effects of melatonin (0, 75, and 150 μM) and salt stress (0, 50 and 100 mM NaCl) were investigated by assaying the some agronomic, physlogical and biochemical attributes and essential oil compounds of bitter melon (Momordica charantia). The results showed that exogenous melatonin could promote net photosynthetic rate (Pn) and PSII efficiency (Fv/Fm), increase K+ content and activity of antioxidant enzymes and decrease reactive oxygen species, malondialdehyde and Na+ content in stress-submitted seedlings, in comparison to the non-stressed seedlings (p < 0.05). Melatonin increased content of essential oils. Concerning the major compounds of fruits of bitter melon, charantin, momordicin and cucurbitacin were increased with the melatonin treatments, whereas they were critically decreased with the salt stress. In addition, melatonin increased the antioxidant capacity in fruits under non-saline and salinity conditions. Amid the concentrations of melatonin, plants treated with 150 μM of melatonin under either non-saline or saline conditions showed better performance and productivity. Therefore, application of 150 μM melatonin resulted in a significant improvement of salinity tolerance and essential oil compounds in bitter melon plant, suggesting this as an efficient 'green' strategy for sustainable crop production under salt stress conditions.
Collapse
Affiliation(s)
- Morteza Sheikhalipour
- Department of Horticulture, Faculty of Horticulture, University of Mohaghegh Ardabili, Ardabil, Iran; Department of Plant Breeding and Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Gholamreza Gohari
- Department of Horticultural Science, Faculty of Agriculture, University of Maragheh, Maragheh, Iran; Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology Limassol, Cyprus.
| | - Behrooz Esmaielpour
- Department of Horticulture, Faculty of Horticulture, University of Mohaghegh Ardabili, Ardabil, Iran.
| | - Mehdi Behnamian
- Department of Horticulture, Faculty of Horticulture, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mousa Torabi Giglou
- Department of Horticulture, Faculty of Horticulture, University of Mohaghegh Ardabili, Ardabil, Iran
| | | | | | - Muhittin Kulak
- Department of Herbal and Animal Production, Vocational School of Technical Sciences, Igdir University, Igdir, Turkey
| | - Andreas Ioannou
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology Limassol, Cyprus
| | - Vasileios Fotopoulos
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology Limassol, Cyprus
| | - Federico Vita
- Department of Biology, University of Bari Aldo Moro, 70126, Bari, Italy
| |
Collapse
|
10
|
Yang YX, Wang XY, Lin T, Sun Y, Yu YC, Zhu ZH. Opportunities and challenges for ribosome-inactivating proteins in traditional Chinese medicine plants. Toxicon 2023; 234:107278. [PMID: 37683701 DOI: 10.1016/j.toxicon.2023.107278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023]
Abstract
Ribosome-inactivating proteins (RIPs) are a class of cytotoxic rRNA N-glycosylase, which widely exist in higher plants in different taxonomy, including many traditional Chinese medicinal materials and vegetables and fruits. In this paper, the traditional Chinese medicinal plants containing RIPs protein were sorted out, and their pharmacological effects and clinical applications were analyzed. Since many RIPs in traditional Chinese medicine plants exhibit antiviral and antitumor activities and show great clinical application potential, people's interest in these proteins is on the rise. This paper summarizes the possible mechanism of RIPs's anti-virus and anti-tumor effects, and discusses its potential problems and risks, laying a foundation for subsequent research on how to exert its anti-virus and anti-tumor effects.
Collapse
Affiliation(s)
- Yi-Xuan Yang
- School of Life Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Xin-Yi Wang
- School of Life Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Tong Lin
- School of Life Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Yu Sun
- School of Life Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Yi-Cheng Yu
- School of Life Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Zhen-Hong Zhu
- School of Life Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
11
|
Safaei S, Imani M. Computational design of a chimeric toxin against Claudin-4-expressing cancer cells: molecular modeling, docking and molecular dynamics simulation analysis. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2023; 14:259-265. [PMID: 37342289 PMCID: PMC10278906 DOI: 10.30466/vrf.2022.548415.3378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/11/2022] [Indexed: 06/22/2023]
Abstract
Cancer is one of the main reasons of mortality all over the world. Over the time, the major ways for cancer-therapy were based on radiotherapy, chemotherapy and surgery. These methods are not specific enough for that purpose, therefore, new ideas for design of new drugs with higher specificity are considered. Chimeric protein toxins are hybrid proteins consisting of a targeting portion and a toxic one which specifically bind and kill the target cancer cells. The main purpose of this study was designing a recombinant chimeric toxin with biding capability to one of the most key receptors namely claudin-4 which is over-expressed in almost all cancer cells. To design it, we utilized the last 30 C-terminal amino acids of Clostridium perfringens enterotoxin (CPE) as a binding module for claudin-4 and the toxic module which is the A-domain of Shiga toxin from Shigella dysenteriae. Using molecular modeling and docking methods, appropriate binding affinity of the recombinant chimeric toxin to its specific receptor was demonstrated. In the next step, the stability of this interaction was investigated by molecular dynamics simulation. Although partial instability was detected at some time points, however, sufficient stable situation of hydrogens bonds and high binding affinity between the chimeric toxin and receptor were observed in the in silico studies which in turn suggested that this complex could be formed successfully.
Collapse
Affiliation(s)
| | - Mehdi Imani
- Correspondence:Mehdi Imani. Msc, PhD, Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran, E-mail:
| |
Collapse
|
12
|
Bortolotti M, Biscotti F, Zanello A, Bolognesi A, Polito L. New Insights on Saporin Resistance to Chemical Derivatization with Heterobifunctional Reagents. Biomedicines 2023; 11:biomedicines11041214. [PMID: 37189832 DOI: 10.3390/biomedicines11041214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Saporin is a type 1 ribosome-inactivating protein widely used as toxic payload in the construction of targeted toxins, chimeric molecules formed by a toxic portion linked to a carrier moiety. Among the most used carriers, there are large molecules (mainly antibodies) and small molecules (such as neurotransmitters, growth factors and peptides). Some saporin-containing targeted toxins have been used for the experimental treatment of several diseases, giving very promising results. In this context, one of the reasons for the successful use of saporin lies in its resistance to proteolytic enzymes and to conjugation procedures. In this paper, we evaluated the influence of derivatization on saporin using three heterobifunctional reagents, namely 2-iminothiolane (2-IT), N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP) and 4-succinimidyloxycarbonyl-α-methyl-α-[2-pyridyldithio]toluene (SMPT). In order to obtain the highest number of inserted -SH groups with the lowest reduction of saporin biological activities, we assessed the residual ability of saporin to inhibit protein synthesis, to depurinate DNA and to induce cytotoxicity after derivatization. Our results demonstrate that saporin maintains an excellent resistance to derivatization processes, especially with SPDP, and permit us to define reaction conditions, in which saporin biological properties may not be altered. Therefore, these findings provide useful information for the construction of saporin-based targeted toxins, especially with small carriers.
Collapse
Affiliation(s)
- Massimo Bortolotti
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Francesco Biscotti
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Andrea Zanello
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Andrea Bolognesi
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Letizia Polito
- Department of Medical and Surgical Sciences-DIMEC, General Pathology Section, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
13
|
Citores L, Ferreras JM. Biological Activities of Ribosome-Inactivating Proteins. Toxins (Basel) 2023; 15:35. [PMID: 36668855 PMCID: PMC9865172 DOI: 10.3390/toxins15010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
After more than 50 years of research, studies on the structure and biological activities of ribosome-inactivating proteins (RIPs) continue to provide a field of great interest within the scientific community, both for the health risks they pose and their applications in medicine and biotechnology [...].
Collapse
Affiliation(s)
- Lucía Citores
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, 47011 Valladolid, Spain
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, 47011 Valladolid, Spain
| |
Collapse
|
14
|
Muribeca ADJB, Gomes PWP, Paes SS, da Costa APA, Gomes PWP, Viana JDS, Reis JDE, Pamplona SDGSR, Silva C, Bauermeister A, Santos LDS, da Silva MN. Antibacterial Activity from Momordica charantia L. Leaves and Flavones Enriched Phase. Pharmaceutics 2022; 14:1796. [PMID: 36145544 PMCID: PMC9505480 DOI: 10.3390/pharmaceutics14091796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/23/2022] Open
Abstract
Momordica charantia L. (Cucurbitaceae) is a plant known in Brazil as "melão de São Caetano", which has been related to many therapeutic applications in folk medicine. Herein, we describe antibacterial activities and related metabolites for an extract and fractions obtained from the leaves of that species. An ethanolic extract and its three fractions were used to perform in vitro antibacterial assays. In addition, liquid chromatography coupled to mass spectrometry and the molecular networking approach were used for the metabolite annotation process. Overall, 25 compounds were annotated in the ethanolic extract from M. charantia leaves, including flavones, terpenes, organic acids, and inositol pyrophosphate derivatives. The ethanolic extract exhibited low activity against Proteus mirabilis (MIC 312.5 µg·mL-1) and Klebsiella pneumoniae (MIC 625 µg·mL-1). The ethyl acetate phase showed interesting antibacterial activity (MIC 156.2 µg·mL-1) against Klebsiella pneumoniae, and it was well justified by the high content of glycosylated flavones. Therefore, based on the ethyl acetate phase antibacterial result, we suggest that M. charantia leaves could be considered as an alternative antibacterial source against K. pneumoniae and can serve as a pillar for future studies as well as pharmacological application against the bacteria.
Collapse
Affiliation(s)
- Abraão de Jesus B. Muribeca
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
| | - Paulo Wender P. Gomes
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, San Diego, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| | - Steven Souza Paes
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
| | - Ana Paula Alves da Costa
- Department of Natural Science, Campus XIX, State University of Pará, Rodovia PA 154, Km 28, Cajú, Salvaterra 66860-000, PA, Brazil
| | - Paulo Weslem Portal Gomes
- Institute of Biology, University of Campinas, Monteiro Lobato, 255, Barão Geraldo, Campinas 13083-862, SP, Brazil
| | - Jéssica de Souza Viana
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
| | - José Diogo E. Reis
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
| | | | - Consuelo Silva
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
- Pharmaceutical Science Post-Graduation Program, Faculty of Pharmacy, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Anelize Bauermeister
- Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil
| | - Lourivaldo da Silva Santos
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
| | - Milton Nascimento da Silva
- Institute of Exact and Natural Sciences, Federal University of Pará, Augusto Corrêa, 01, Guamá, Belém 66075-110, PA, Brazil
| |
Collapse
|
15
|
Zanello A, Bortolotti M, Maiello S, Bolognesi A, Polito L. Anti-PD-L1 immunoconjugates for cancer therapy: Are available antibodies good carriers for toxic payload delivering? Front Pharmacol 2022; 13:972046. [PMID: 36052121 PMCID: PMC9424723 DOI: 10.3389/fphar.2022.972046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/19/2022] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint mechanisms are important molecular cell systems that maintain tolerance toward autoantigens in order to prevent immunity-mediated accidental damage. It is well known that cancer cells may exploit these molecular and cellular mechanisms to escape recognition and elimination by immune cells. Programmed cell death protein-1 (PD-1) and its natural ligand programmed cell death ligand-1 (PD-L1) form the PD-L1/PD-1 axis, a well-known immune checkpoint mechanism, which is considered an interesting target in cancer immunotherapy. In fact, the expression of PD-L1 was found in various solid malignancies and the overactivation of PD-L1/PD-1 axis results in a poor patient survival rate. Breaking PD-L1/PD-1 axis, by blocking either the cancer side or the immune side of the axis, is currently used as anti-cancer strategy to re-establish a tumor-specific immune response. For this purpose, several blocking antibodies are now available. To date, three anti-PD-L1 antibodies have been approved by the FDA, namely atezolizumab, durvalumab and avelumab. The main advantages of anti-PD-L1 antibodies arise from the overexpression of PD-L1 antigen by a high number of tumor cells, also deriving from different tissues; this makes anti-PD-L1 antibodies potential pan-specific anti-cancer molecules. Despite the good results reported in clinical trials with anti-PD-L1 antibodies, there is a significant number of patients that do not respond to the therapy. In fact, it should be considered that, in some neoplastic patients, reduced or absent infiltration of cytotoxic T cells and natural killer cells in the tumor microenvironment or presence of other immunosuppressive molecules make immunotherapy with anti-PD-L1 blocking antibodies less effective. A strategy to improve the efficacy of antibodies is to use them as carriers for toxic payloads (toxins, drugs, enzymes, radionuclides, etc.) to form immunoconjugates. Several immunoconjugates have been already approved by FDA for treatment of malignancies. In this review, we focused on PD-L1 targeting antibodies utilized as carrier to construct immunoconjugates for the potential elimination of neoplastic cells, expressing PD-L1. A complete examination of the literature regarding anti-PD-L1 immunoconjugates is here reported, describing the results obtained in vitro and in vivo. The real potential of anti-PD-L1 antibodies as carriers for toxic payload delivery is considered and extensively discussed.
Collapse
|
16
|
Nassiri M, Gopalan V, Vakili-Azghandi M. Modifications of Ribonucleases in Order to Enhance Cytotoxicity in Anticancer Therapy. Curr Cancer Drug Targets 2022; 22:373-387. [PMID: 35240973 DOI: 10.2174/1568009622666220303101005] [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: 08/20/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
Ribonucleases (RNases) are a superfamily of enzymes that have been extensively studied since the 1960s. For a long time, this group of secretory enzymes was studied as an important model for protein chemistry such as folding, stability and enzymatic catalysis. Since it was discovered that RNases displayed cytotoxic activity against several types of malignant cells, recent investigation has focused mainly on the biological functions and medical applications of engineered RNases. In this review, we describe structures, functions and mechanisms of antitumor activity of RNases. They operate at the crossroads of transcription and translation, preferentially degrading tRNA. As a result, this inhibits protein synthesis, induces apoptosis and causes death of cancer cells. This effect can be enhanced thousands of times when RNases are conjugated with monoclonal antibodies. Such combinations, called immunoRNases, have demonstrated selective antitumor activity against cancer cells both in vitro and in animal models. This review summarizes the current status of engineered RNases and immunoRNases as promising novel therapeutic agents for different types of cancer. Also, we describe our experimental results from published or previously unpublished research and compare with other scientific information.
Collapse
Affiliation(s)
- Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
- School of Life and Environmental Sciences, The University of Sydney, Sydney 2006, NSW, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Queensland 4222, Australia
| | | |
Collapse
|
17
|
Polito L. Antibody Based Delivery of Toxins and Other Active Molecules for Cancer Therapy. Biomedicines 2022; 10:biomedicines10020267. [PMID: 35203476 PMCID: PMC8869543 DOI: 10.3390/biomedicines10020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum-University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy
| |
Collapse
|
18
|
Toxin and Immunotoxin Based Therapeutic Approaches. Toxins (Basel) 2022; 14:toxins14010063. [PMID: 35051040 PMCID: PMC8778363 DOI: 10.3390/toxins14010063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
|
19
|
Lu J, Ding J, Liu Z, Chen T. Retrospective analysis of the preparation and application of immunotherapy in cancer treatment (Review). Int J Oncol 2022; 60:12. [PMID: 34981814 PMCID: PMC8759346 DOI: 10.3892/ijo.2022.5302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibody technology plays a vital role in biomedical and immunotherapy, which greatly promotes the study of the structure and function of genes and proteins. To date, monoclonal antibodies have gone through four stages: murine monoclonal antibody, chimeric monoclonal antibody, humanised monoclonal antibody and fully human monoclonal antibody; thousands of monoclonal antibodies have been used in the fields of biology and medicine, playing a special role in the pathogenesis, diagnosis and treatment of disease. In this review, we compare the advantages and disadvantages of hybridoma technology, phage display technology, ribosome display technology, transgenic mouse technology, single B cell monoclonal antibody generation technologies, and forecast the promising applications of these technologies in clinical medicine, disease diagnosis and tumour treatment.
Collapse
Affiliation(s)
- Jiachen Lu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianing Ding
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
20
|
Wang S, Liu Q, Zeng T, Zhan J, Zhao H, Ho CT, Xiao Y, Li S. Immunomodulatory effects and associated mechanisms of Momordica charantia and its phytochemicals. Food Funct 2022; 13:11986-11998. [DOI: 10.1039/d2fo02096c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Momordica charantia L. (M. charantia), which is a member of the Cucurbitaceae family and widely distributed in tropical and subtropical regions, has been consumed as a vegetable and also used as herbal medicine for thousands of years worldwide.
Collapse
Affiliation(s)
- Shuzhen Wang
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, Shandong Province, P.R. China
| | - Ting Zeng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 250355, Shandong Province, P.R. China
| | - Jianfeng Zhan
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Yunli Xiao
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
| | - Shiming Li
- College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, 438000, Hubei Province, P.R. China
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
21
|
Maiello S, Iglesias R, Polito L, Citores L, Bortolotti M, Ferreras JM, Bolognesi A. Sequence, Structure, and Binding Site Analysis of Kirkiin in Comparison with Ricin and Other Type 2 RIPs. Toxins (Basel) 2021; 13:toxins13120862. [PMID: 34941700 PMCID: PMC8705660 DOI: 10.3390/toxins13120862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/29/2022] Open
Abstract
Kirkiin is a new type 2 ribosome-inactivating protein (RIP) purified from the caudex of Adenia kirkii with a cytotoxicity compared to that of stenodactylin. The high toxicity of RIPs from Adenia genus plants makes them interesting tools for biotechnology and therapeutic applications, particularly in cancer therapy. The complete amino acid sequence and 3D structure prediction of kirkiin are here reported. Gene sequence analysis revealed that kirkiin is encoded by a 1572 bp open reading frame, corresponding to 524 amino acid residues, without introns. The amino acid sequence analysis showed a high degree of identity with other Adenia RIPs. The 3D structure of kirkiin preserves the overall folding of type 2 RIPs. The key amino acids of the active site, described for ricin and other RIPs, are also conserved in the kirkiin A chain. Sugar affinity studies and docking experiments revealed that both the 1α and 2γ sites of the kirkiin B chain exhibit binding activity toward lactose and D-galactose, being lower than ricin. The replacement of His246 in the kirkiin 2γ site instead of Tyr248 in ricin causes a different structure arrangement that could explain the lower sugar affinity of kirkiin with respect to ricin.
Collapse
Affiliation(s)
- Stefania Maiello
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (S.M.); (M.B.); (A.B.)
| | - Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, 47011 Valladolid, Spain; (L.C.); (J.M.F.)
- Correspondence: (R.I.); (L.P.)
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (S.M.); (M.B.); (A.B.)
- Correspondence: (R.I.); (L.P.)
| | - Lucía Citores
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, 47011 Valladolid, Spain; (L.C.); (J.M.F.)
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (S.M.); (M.B.); (A.B.)
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, 47011 Valladolid, Spain; (L.C.); (J.M.F.)
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (S.M.); (M.B.); (A.B.)
| |
Collapse
|
22
|
Wong JJW, Selbo PK. Light-controlled elimination of PD-L1+ cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 225:112355. [PMID: 34768077 DOI: 10.1016/j.jphotobiol.2021.112355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/11/2021] [Accepted: 11/03/2021] [Indexed: 12/31/2022]
Abstract
The programmed death ligand-1 (PD-L1), also known as CD274 or B7-H1, is mainly expressed on cancer cells and/or immunosuppressive cells in the tumor microenvironment (TME) and plays an essential role in tumor progression and immune escape. Immune checkpoint inhibitors (ICIs) of the PD-1/PD-L1 axis have shown impressive clinical success, however, the majority of the patients do not respond to immune checkpoint therapy (ICT). Thus, to overcome ICT resistance there is a high need for potent and novel strategies that simultaneously target both tumor cells and immunosuppressive cells in the TME. In this study, we show that the intracellular light-controlled drug delivery method photochemical internalization (PCI) induce specific and strongly enhanced cytotoxic effects of the PD-L1-targeting immunotoxin, anti-PD-L1-saporin (Anti-PDL1-SAP), in the PD-L1+ triple-negative breast cancer MDA-MB-231 cell line, while no enhanced efficacy was obtained in the PD-L1 negative control cell line MDA-MB-453. Using fluorescence microscopy, we reveal that the anti-PD-L1 antibody binds to PD-L1 on the surface of the MDA-MD-231 cells and overnight accumulates in late endosomes and lysosomes where it co-localizes with the PCI photosensitizer fimaporfin (TPCS2a). Moreover, light-controlled endosomal/lysosomal escape of the anti-PD-L1 antibody and fimaporfin into the cytosol was obtained. We also confirm that the breast MDA-MB-468 and the prostate PC-3 and DU-145 cancer cell lines have subpopulations with PD-L1 expression. In addition, we show that interferon-gamma strongly induce PD-L1 expression in the per se PD-L1 negative CT26.WT cells and enhance the PD-L1 expression in MC-38 cells, of which both are murine colon cancer cell lines. In conclusion, our work provides an in vitro proof-of-concept of PCI-enhanced targeting and eradication of PD-L1 positive immunosuppressive cells. This light-controlled combinatorial strategy has a potential to advance cancer immunotherapy and should be explored in preclinical studies.
Collapse
Affiliation(s)
- Judith Jing Wen Wong
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway.
| | - Pål Kristian Selbo
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway.
| |
Collapse
|
23
|
Li XP, Harijan RK, Cao B, Kahn JN, Pierce M, Tsymbal AM, Roberge JY, Augeri D, Tumer NE. Synthesis and Structural Characterization of Ricin Inhibitors Targeting Ribosome Binding Using Fragment-Based Methods and Structure-Based Design. J Med Chem 2021; 64:15334-15348. [PMID: 34648707 PMCID: PMC10704857 DOI: 10.1021/acs.jmedchem.1c01370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ricin toxin A subunit (RTA) is the catalytic subunit of ricin, which depurinates an adenine from the sarcin/ricin loop in eukaryotic ribosomes. There are no approved inhibitors against ricin. We used a new strategy to disrupt RTA-ribosome interactions by fragment screening using surface plasmon resonance. Here, using a structure-guided approach, we improved the affinity and inhibitory activity of small-molecular-weight lead compounds and obtained improved compounds with over an order of magnitude higher efficiency. Four advanced compounds were characterized by X-ray crystallography. They bind at the RTA-ribosome binding site as the original compound but in a distinctive manner. These inhibitors bind remotely from the catalytic site and cause local conformational changes with no alteration of the catalytic site geometry. Yet they inhibit depurination by ricin holotoxin and inhibit the cytotoxicity of ricin in mammalian cells. They are the first agents that protect against ricin holotoxin by acting directly on RTA.
Collapse
Affiliation(s)
- Xiao-Ping Li
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| | - Rajesh K Harijan
- Department of Biochemistry, Albert Einstein College of Medicine, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Bin Cao
- Molecular Design and Synthesis Core, Rutgers University Biomolecular Innovations Cores, Office for Research, Rutgers University, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| | - Jennifer N Kahn
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| | - Michael Pierce
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| | - Anastasiia M Tsymbal
- Molecular Design and Synthesis Core, Rutgers University Biomolecular Innovations Cores, Office for Research, Rutgers University, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| | - Jacques Y Roberge
- Molecular Design and Synthesis Core, Rutgers University Biomolecular Innovations Cores, Office for Research, Rutgers University, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| | - David Augeri
- Molecular Design and Synthesis Core, Rutgers University Biomolecular Innovations Cores, Office for Research, Rutgers University, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| | - Nilgun E Tumer
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| |
Collapse
|
24
|
Khirehgesh MR, Sharifi J, Safari F, Akbari B. Immunotoxins and nanobody-based immunotoxins: review and update. J Drug Target 2021; 29:848-862. [PMID: 33615933 DOI: 10.1080/1061186x.2021.1894435] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunotoxins (ITs) are protein-based drugs that compose of targeting and cytotoxic moieties. After binding the IT to the specific cell-surface antigen, the IT internalises into the target cell and kills it. Targeting and cytotoxic moieties usually include monoclonal antibodies and protein toxins with bacterial or plant origin, respectively. ITs have been successful in haematologic malignancies treatment. However, ITs penetrate poorly into solid tumours because of their large size. Use of camelid antibody fragments known as nanobodies (Nbs) as a targeting moiety may overcome this problem. Nbs are the smallest fragment of antibodies with excellent tumour tissue penetration. The ability to recognise cryptic (immuno-evasive) target antigens, low immunogenicity, and high-affinity are other fundamental characteristics of Nbs that make them suitable candidates in targeted therapy. Here, we reviewed and discussed the structure and function of ITs, Nbs, and nanobody-based ITs. To gain sound insight into the issue at hand, we focussed on nanobody-based ITs.
Collapse
Affiliation(s)
- Mohammad Reza Khirehgesh
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jafar Sharifi
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Akbari
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
25
|
Antibody Conjugates for Sarcoma Therapy: How Far along Are We? Biomedicines 2021; 9:biomedicines9080978. [PMID: 34440182 PMCID: PMC8392509 DOI: 10.3390/biomedicines9080978] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 01/12/2023] Open
Abstract
Sarcomas are one of the most difficult type of cancer to manage and treat because of their extremely heterogeneous molecular and morphological features. Despite the progress made over the years in the establishment of standard protocols for high and low grading/staging sarcoma patients, mostly with chemotherapy and/or radiotherapy, 50% of treated patients experience relapse episodes. Because of this, in the last 20 years, new therapeutic approaches for sarcoma treatment have been evaluated in preclinical and clinical studies. Among them, antibody-based therapies have been the most studied. Immunoconjugates consist of a carrier portion, frequently represented by an antibody, linked to a toxic moiety, i.e., a drug, toxin, or radionuclide. While the efficacy of immunoconjugates is well demonstrated in the therapy of hematological tumors and more recently also of epithelial ones, their potential as therapeutic agents against sarcomas is still not completely explored. In this paper, we summarize the results obtained with immunoconjugates targeting sarcoma surface antigens, considering both preclinical and clinical studies. To date, the encouraging results obtained in preclinical studies allowed nine immunoconjugates to enter clinical trials, demonstrating the validity of immunotherapy as a promising pharmacological tool also for sarcoma therapy.
Collapse
|
26
|
Yang S, Ngai WSC, Chen PR. Chemical engineering of bacterial effectors for regulating cell signaling and responses. Curr Opin Chem Biol 2021; 64:48-56. [PMID: 33993047 DOI: 10.1016/j.cbpa.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/31/2021] [Accepted: 04/10/2021] [Indexed: 01/24/2023]
Abstract
Bacteria have evolved a variety of effector proteins to facilitate their survival and proliferation within the host environment. Continuous competition at the host-pathogen interface has empowered these effectors with unique mechanism and high specificity toward their host targets. The rich repertoire of bacterial effectors has thus provided us an attractive toolkit for investigating various cellular processes, such as signal transductions. With recent advances in protein chemistry and engineering, we now have the capability for on-demand control of protein activity with high precision. Herein, we review the development of chemically engineered bacterial effectors to control kinase-mediated signal transductions, inhibit protein translation, and direct genetic editing within host cells. We also highlight future opportunities for harnessing diverse prokaryotic effectors as powerful tools for mechanistic investigation and therapeutic intervention of eukaryotic systems.
Collapse
Affiliation(s)
- Shaojun Yang
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - William Shu Ching Ngai
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Peng R Chen
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
27
|
Polito L, Bortolotti M, Iglesias R, Bolognesi A. Editorial: Toxic Plant Proteins as Experimental Drugs for Human Pathologies. Front Pharmacol 2021; 12:689924. [PMID: 33995114 PMCID: PMC8113869 DOI: 10.3389/fphar.2021.689924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, Valladolid, Spain
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
28
|
Desai SA, Manjappa A, Khulbe P. Drug delivery nanocarriers and recent advances ventured to improve therapeutic efficacy against osteosarcoma: an overview. J Egypt Natl Canc Inst 2021; 33:4. [PMID: 33555490 DOI: 10.1186/s43046-021-00059-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/18/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is one of the key cancers affecting the bone tissues, primarily occurred in children and adolescence. Recently, chemotherapy followed by surgery and then post-operative adjuvant chemotherapy is widely used for the treatment of OS. However, the lack of selectivity and sensitivity to tumor cells, the development of multi-drug resistance (MDR), and dangerous side effects have restricted the use of chemotherapeutics. MAIN BODY There is an unmet need for novel drug delivery strategies for effective treatment and management of OS. Advances in nanotechnology have led to momentous progress in the design of tumor-targeted drug delivery nanocarriers (NCs) as well as functionalized smart NCs to achieve targeting and to treat OS effectively. The present review summarizes the drug delivery challenges in OS, and how organic nanoparticulate approaches are useful in overcoming barriers will be explained. The present review describes the various organic nanoparticulate approaches such as conventional nanocarriers, stimuli-responsive NCs, and ligand-based active targeting strategies tested against OS. The drug conjugates prepared with copolymer and ligand having bone affinity, and advanced promising approaches such as gene therapy, gene-directed enzyme prodrug therapy, and T cell therapy tested against OS along with their reported limitations are also briefed in this review. CONCLUSION The nanoparticulate drugs, drug conjugates, and advanced therapies such as gene therapy, and T cell therapy have promising and potential application in the effective treatment of OS. However, many of the above approaches are still at the preclinical stage, and there is a long transitional period before their clinical application.
Collapse
Affiliation(s)
- Sujit Arun Desai
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Rd, Mahal, Jagatpura, Jaipur, Rajasthan, 302017, India. .,Annasaheb Dange College of D Pharmacy, Ashta, Tal: Walwa, Dist., Sangli, Maharashtra, 416301, India.
| | - Arehalli Manjappa
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist., Kolhapur, Maharashtra, 416113, India
| | - Preeti Khulbe
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Rd, Mahal, Jagatpura, Jaipur, Rajasthan, 302017, India
| |
Collapse
|
29
|
Iglesias R, Ferreras JM, Llorente A, Citores L. Ebulin l Is Internalized in Cells by Both Clathrin-Dependent and -Independent Mechanisms and Does Not Require Clathrin or Dynamin for Intoxication. Toxins (Basel) 2021; 13:toxins13020102. [PMID: 33573355 PMCID: PMC7911328 DOI: 10.3390/toxins13020102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/12/2021] [Accepted: 01/27/2021] [Indexed: 11/21/2022] Open
Abstract
Ebulin l is an A-B toxin, and despite the presence of a B chain, this toxin displays much less toxicity to cells than the potent A-B toxin ricin. Here, we studied the binding, mechanisms of endocytosis, and intracellular pathway followed by ebulin l and compared it with ricin. COS-1 cells and HeLa cells with inducible synthesis of a mutant dynamin (K44A) were used in this study. The transport of these toxins was measured using radioactively or fluorescently labeled toxins. The data show that ebulin l binds to cells to a lesser extent than ricin. Moreover, the expression of mutant dynamin does not affect the endocytosis, degradation, or toxicity of ebulin l. However, the inhibition of clathrin-coated pit formation by acidification of the cytosol reduced ebulin l endocytosis but not toxicity. Remarkably, unlike ricin, ebulin l is not transported through the Golgi apparatus to intoxicate the cells and ebulin l induces apoptosis as the predominant cell death mechanism. Therefore, after binding to cells, ebulin l is taken up by clathrin-dependent and -independent endocytosis into the endosomal/lysosomal system, but there is no apparent role for clathrin and dynamin in productive intracellular routing leading to intoxication.
Collapse
Affiliation(s)
- Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (J.M.F.)
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (J.M.F.)
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway;
- Department of Mechanical, Electronics and Chemical Engineering Art and Design, Oslo Metropolitan University, 0130 Oslo, Norway
| | - Lucía Citores
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (J.M.F.)
- Correspondence:
| |
Collapse
|
30
|
Bortolotti M, Maiello S, Ferreras JM, Iglesias R, Polito L, Bolognesi A. Kirkiin: A New Toxic Type 2 Ribosome-Inactivating Protein from the Caudex of Adenia kirkii. Toxins (Basel) 2021; 13:toxins13020081. [PMID: 33499082 PMCID: PMC7912562 DOI: 10.3390/toxins13020081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022] Open
Abstract
Ribosome-inactivating proteins (RIPs) are plant toxins that irreversibly damage ribosomes and other substrates, thus causing cell death. RIPs are classified in type 1 RIPs, single-chain enzymatic proteins, and type 2 RIPs, consisting of active A chains, similar to type 1 RIPs, linked to lectin B chains, which enable the rapid internalization of the toxin into the cell. For this reason, many type 2 RIPs are very cytotoxic, ricin, volkensin and stenodactylin being the most toxic ones. From the caudex of Adenia kirkii (Mast.) Engl., a new type 2 RIP, named kirkiin, was purified by affinity chromatography on acid-treated Sepharose CL-6B and gel filtration. The lectin, with molecular weight of about 58 kDa, agglutinated erythrocytes and inhibited protein synthesis in a cell-free system at very low concentrations. Moreover, kirkiin was able to depurinate mammalian and yeast ribosomes, but it showed little or no activity on other nucleotide substrates. In neuroblastoma cells, kirkiin inhibited protein synthesis and induced apoptosis at doses in the pM range. The biological characteristics of kirkiin make this protein a potential candidate for several experimental pharmacological applications both alone for local treatments and as component of immunoconjugates for systemic targeting in neurodegenerative studies and cancer therapy.
Collapse
Affiliation(s)
- Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy; (M.B.); (S.M.); (A.B.)
| | - Stefania Maiello
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy; (M.B.); (S.M.); (A.B.)
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (J.M.F.); (R.I.)
| | - Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (J.M.F.); (R.I.)
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy; (M.B.); (S.M.); (A.B.)
- Correspondence:
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy; (M.B.); (S.M.); (A.B.)
| |
Collapse
|
31
|
Cai Y, Yu S, Chi X, Radoshitzky SR, Kuhn JH, Berger EA. An immunotoxin targeting Ebola virus glycoprotein inhibits Ebola virus production from infected cells. PLoS One 2021; 16:e0245024. [PMID: 33411835 PMCID: PMC7790382 DOI: 10.1371/journal.pone.0245024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/19/2020] [Indexed: 12/29/2022] Open
Abstract
Ebola virus (EBOV), a member of the mononegaviral family Filoviridae, causes severe disease associated with high lethality in humans. Despite enormous progress in development of EBOV medical countermeasures, no anti-EBOV treatment has been approved. We designed an immunotoxin in which a single-chain variable region fragment of the EBOV glycoprotein-specific monoclonal antibody 6D8 was fused to the effector domains of Pseudomonas aeruginosa exotoxin A (PE38). This immunotoxin, 6D8-PE38, bound specifically to cells expressing EBOV glycoproteins. Importantly, 6D8-PE38 targeted EBOV-infected cells, as evidenced by inhibition of infectious EBOV production from infected cells, including primary human macrophages. The data presented here provide a proof of concept for immunotoxin-based targeted killing of infected cells as a potential antiviral intervention for Ebola virus disease.
Collapse
Affiliation(s)
- Yingyun Cai
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, United States of America
| | - Shuiqing Yu
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, United States of America
| | - Xiaoli Chi
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Sheli R. Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, United States of America
| | - Edward A. Berger
- Laboratory of Viral Disease, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
32
|
Iglesias R, Polito L, Bortolotti M, Pedrazzi M, Citores L, Ferreras JM, Bolognesi A. Primary Sequence and 3D Structure Prediction of the Plant Toxin Stenodactylin. Toxins (Basel) 2020; 12:toxins12090538. [PMID: 32825611 PMCID: PMC7551084 DOI: 10.3390/toxins12090538] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 01/04/2023] Open
Abstract
Stenodactylin is one of the most potent type 2 ribosome-inactivating proteins (RIPs); its high toxicity has been demonstrated in several models both in vitro and in vivo. Due to its peculiarities, stenodactylin could have several medical and biotechnological applications in neuroscience and cancer treatment. In this work, we report the complete amino acid sequence of stenodactylin and 3D structure prediction. The comparison between the primary sequence of stenodactylin and other RIPs allowed us to identify homologies/differences and the amino acids involved in RIP toxic activity. Stenodactylin RNA was isolated from plant caudex, reverse transcribed through PCR and the cDNA was amplificated and cloned into a plasmid vector and further analyzed by sequencing. Nucleotide sequence analysis showed that stenodactylin A and B chains contain 251 and 258 amino acids, respectively. The key amino acids of the active site described for ricin and most other RIPs are also conserved in the stenodactylin A chain. Stenodactylin amino acid sequence shows a high identity degree with volkensin (81.7% for A chain, 90.3% for B chain), whilst when compared with other type 2 RIPs the identity degree ranges from 27.7 to 33.0% for the A chain and from 42.1 to 47.7% for the B chain.
Collapse
Affiliation(s)
- Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E−47011 Valladolid, Spain; (R.I.); (L.C.)
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (L.P.); (M.B.); (M.P.)
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (L.P.); (M.B.); (M.P.)
| | - Manuela Pedrazzi
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (L.P.); (M.B.); (M.P.)
| | - Lucía Citores
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E−47011 Valladolid, Spain; (R.I.); (L.C.)
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E−47011 Valladolid, Spain; (R.I.); (L.C.)
- Correspondence: (J.M.F.); (A.B.)
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine—DIMES, General Pathology Section, Alma Mater Studiorum—University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy; (L.P.); (M.B.); (M.P.)
- Correspondence: (J.M.F.); (A.B.)
| |
Collapse
|
33
|
Antiviral Activity of PD-L1 and PD-L4, Type 1 Ribosome Inactivating Proteins from Leaves of Phytolacca dioica L. in the Pathosystem Phaseolus vulgaris-Tobacco Necrosis Virus (TNV). Toxins (Basel) 2020; 12:toxins12080524. [PMID: 32824023 PMCID: PMC7472211 DOI: 10.3390/toxins12080524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Using the pathosystem Phaseolus vulgaris-tobacco necrosis virus (TNV), we demonstrated that PD-L1 and PD-L4, type-1 ribosome inactivating proteins (RIPs) from leaves of Phytolacca dioica L., possess a strong antiviral activity. This activity was exerted both when the RIPs and the virus were inoculated together in the same leaf and when they were inoculated or applied separately in the adaxial and abaxial leaf surfaces. This suggests that virus inhibition would mainly occur inside plant cells at the onset of infection. Histochemical studies showed that both PD-L1 and PD-L4 were not able to induce oxidative burst and cell death in treated leaves, which were instead elicited by inoculation of the virus alone. Furthermore, when RIPs and TNV were inoculated together, no sign of H2O2 deposits and cell death were detectable, indicating that the virus could have been inactivated in a very early stage of infection, before the elicitation of a hypersensitivity reaction. In conclusion, the strong antiviral activity is likely exerted inside host cells as soon the virus disassembles to start translation of the viral genome. This activity is likely directed towards both viral and ribosomal RNA, explaining the almost complete abolition of infection when virus and RIP enter together into the cells.
Collapse
|
34
|
Production of Recombinant Gelonin Using an Automated Liquid Chromatography System. Toxins (Basel) 2020; 12:toxins12080519. [PMID: 32823678 PMCID: PMC7472732 DOI: 10.3390/toxins12080519] [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: 07/03/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 11/28/2022] Open
Abstract
Advances in recombinant DNA technology have opened up new possibilities of exploiting toxic proteins for therapeutic purposes. Bringing forth these protein toxins from the bench to the bedside strongly depends on the availability of production methods that are reproducible, scalable and comply with good manufacturing practice (GMP). The type I ribosome-inhibiting protein, gelonin, has great potential as an anticancer drug, but is sequestrated in endosomes and lysosomes. This can be overcome by combination with photochemical internalization (PCI), a method for endosomal drug release. The combination of gelonin-based drugs and PCI represents a tumor-targeted therapy with high precision and efficiency. The aim of this study was to produce recombinant gelonin (rGel) at high purity and quantity using an automated liquid chromatography system. The expression and purification process was documented as highly efficient (4.4 mg gelonin per litre induced culture) and reproducible with minimal loss of target protein (~50% overall yield compared to after initial immobilized metal affinity chromatography (IMAC)). The endotoxin level of 0.05–0.09 EU/mg was compatible with current standards for parenteral drug administration. The automated system provided a consistent output with minimal human intervention and close monitoring of each purification step enabled optimization of both yield and purity of the product. rGel was shown to have equivalent biological activity and cytotoxicity, both with and without PCI-mediated delivery, as rGelref produced without an automated system. This study presents a highly refined and automated manufacturing procedure for recombinant gelonin at a quantity and quality sufficient for preclinical evaluation. The methods established in this report are in compliance with high quality standards and compose a solid platform for preclinical development of gelonin-based drugs.
Collapse
|
35
|
Li XP, Harijan RK, Kahn JN, Schramm VL, Tumer NE. Small Molecule Inhibitors Targeting the Interaction of Ricin Toxin A Subunit with Ribosomes. ACS Infect Dis 2020; 6:1894-1905. [PMID: 32428396 DOI: 10.1021/acsinfecdis.0c00127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Ricin toxin A subunit (RTA) removes an adenine from the universally conserved sarcin/ricin loop (SRL) on eukaryotic ribosomes, thereby inhibiting protein synthesis. No high affinity and selective small molecule therapeutic antidotes have been reported against ricin toxicity. RTA binds to the ribosomal P stalk to access the SRL. The interaction anchors RTA to the P protein C-termini at a well-defined hydrophobic pocket, which is on the opposite face relative to the active site. The RTA ribosome binding site has not been previously targeted by small molecule inhibitors. We used fragment screening with surface plasmon resonance to identify small molecular weight lead compounds that bind RTA and defined their interactions by crystallography. We identified five fragments, which bound RTA with mid-micromolar affinity. Three chemically distinct binding fragments were cocrystallized with RTA, and crystal structures were solved. Two fragments bound at the P stalk binding site, and the third bound to helix D, a motif distinct from the P stalk binding site. All fragments bound RTA remote from the catalytic site and caused little change in catalytic site geometry. Two fragments uniquely bound at the hydrophobic pocket with affinity sufficient to inhibit the catalytic activity on eukaryotic ribosomes in the low micromolar range. The binding mode of these inhibitors mimicked the interaction of the P stalk peptide, establishing that small molecule inhibitors can inhibit RTA binding to the ribosome with the potential for therapeutic intervention.
Collapse
Affiliation(s)
- Xiao-Ping Li
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| | - Rajesh K. Harijan
- Department of Biochemistry, Albert Einstein College of Medicine, Jack and Pearl Resnick Campus,1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Jennifer N. Kahn
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| | - Vern L. Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, Jack and Pearl Resnick Campus,1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Nilgun E. Tumer
- Department of Plant Biology, Rutgers, The State University of New Jersey, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| |
Collapse
|
36
|
Mercatelli D, Bortolotti M, Andresen V, Sulen A, Polito L, Gjertsen BT, Bolognesi A. Early Response to the Plant Toxin Stenodactylin in Acute Myeloid Leukemia Cells Involves Inflammatory and Apoptotic Signaling. Front Pharmacol 2020; 11:630. [PMID: 32457623 PMCID: PMC7226368 DOI: 10.3389/fphar.2020.00630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
Stenodactylin, a highly toxic type 2 ribosome-inactivating protein purified from the caudex of Adenia stenodactyla Harms, is a potential anticancer drug candidate. Previous studies demonstrated that stenodactylin induces apoptosis and necroptosis in treated cells, involving the production of reactive oxygen species. We analyzed the effect of stenodactylin on Raji and Ramos (Human Burkitt’s lymphoma cells) and MOLM-13 (acute myeloid leukemia cells). Moreover, we focused on the early events in MOLM-13 cells that characterize the cellular response to the toxin by whole-genome microarray analysis of gene expression. Treatment with stenodactylin induced the depurination of 28S rRNA within 4 h and increased the phosphorylation of p38 and JNK. A time-dependent activation of caspase 1, 2, 8, 9, 3/7 was also observed. Genome-wide gene expression microarray analysis revealed early changes in the expression of genes involved in the regulation of cell death, inflammation and stress response. After 4 h, a significant increase of transcript level was detectable for ATF3, BTG2, DUSP1, EGR1, and JUN. Increased upstream JUN signaling was also confirmed at protein level. The early response to stenodactylin treatment involves inflammatory and apoptotic signaling compatible with the activation of multiple cell death pathways. Because of the above described properties toward acute myeloid leukemia cells, stenodactylin may be a promising candidate for the design of new immunoconjugates for experimental cancer treatment.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology-FaBiT, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Vibeke Andresen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
| | - André Sulen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Bjørn Tore Gjertsen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
37
|
Lu JQ, Zhu ZN, Zheng YT, Shaw PC. Engineering of Ribosome-inactivating Proteins for Improving Pharmacological Properties. Toxins (Basel) 2020; 12:toxins12030167. [PMID: 32182799 PMCID: PMC7150887 DOI: 10.3390/toxins12030167] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/23/2022] Open
Abstract
Ribosome-inactivating proteins (RIPs) are N-glycosidases, which depurinate a specific adenine residue in the conserved α-sarcin/ricin loop (α-SRL) of rRNA. This loop is important for anchoring elongation factor (EF-G for prokaryote or eEF2 for eukaryote) in mRNA translocation. Translation is inhibited after the attack. RIPs therefore may have been applied for anti-cancer, and anti-virus and other therapeutic applications. The main obstacles of treatment with RIPs include short plasma half-life, non-selective cytotoxicity and antigenicity. This review focuses on the strategies used to improve the pharmacological properties of RIPs on human immunodeficiency virus (HIV) and cancers. Coupling with polyethylene glycol (PEG) increases plasma time and reduces antigenicity. RIPs conjugated with antibodies to form immunotoxins increase the selective toxicity to target cells. The prospects for future development on the engineering of RIPs for improving their pharmacological properties are also discussed.
Collapse
Affiliation(s)
- Jia-Qi Lu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China; (J.-Q.L.); (Z.-N.Z.)
| | - Zhen-Ning Zhu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China; (J.-Q.L.); (Z.-N.Z.)
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms, National Kunming High level Biosafety Research Center for Non-human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China;
| | - Pang-Chui Shaw
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China; (J.-Q.L.); (Z.-N.Z.)
- Correspondence:
| |
Collapse
|
38
|
Ruggiero FM, Rodríguez-Walker M, Daniotti JL. Exploiting the internalization feature of an antibody against the glycosphingolipid SSEA-4 to deliver immunotoxins in breast cancer cells. Immunol Cell Biol 2020; 98:187-202. [PMID: 31916611 DOI: 10.1111/imcb.12314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/16/2019] [Accepted: 01/06/2020] [Indexed: 01/02/2023]
Abstract
The stage-specific embryonic antigen-4 (SSEA-4) is a cell surface glycosphingolipid antigen expressed in early stages of human development. This surface marker is downregulated during the differentiation process but is found re-expressed in several types of tumors, including breast cancer. This feature makes SSEA-4 an attractive target for the development of therapeutic antibodies against tumors. In this work, we first studied the binding and intracellular fate of the monoclonal antibody MC-813-70 directed against SSEA-4. MC-813-70 was found to be rapidly internalized into triple-negative breast cancer cells following binding to its target at the plasma membrane, and to accumulate in acidic organelles, most likely lysosomes. Given the internalization feature of MC-813-70, we next tested whether the antibody was able to selectively deliver the saporin toxin inside SSEA-4-expressing cells. Results show that the immunotoxin complex was properly endocytosed and able to reduce cell viability of breast cancer cells in vitro, either alone or in combination with chemotherapeutic drugs. Our findings indicate that the MC-813-70 antibody has the potential to be developed as an alternative targeted therapeutic agent for cancer cells expressing the SSEA-4 glycolipid.
Collapse
Affiliation(s)
- Fernando M Ruggiero
- Centro de Investigaciones en Química Biológica, de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Macarena Rodríguez-Walker
- Centro de Investigaciones en Química Biológica, de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jose L Daniotti
- Centro de Investigaciones en Química Biológica, de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
39
|
Pierce M, Vengsarkar D, McLaughlin JE, Kahn JN, Tumer NE. Ribosome depurination by ricin leads to inhibition of endoplasmic reticulum stress-induced HAC1 mRNA splicing on the ribosome. J Biol Chem 2019; 294:17848-17862. [PMID: 31624149 DOI: 10.1074/jbc.ra119.009128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/27/2019] [Indexed: 11/06/2022] Open
Abstract
Ricin undergoes retrograde transport to the endoplasmic reticulum (ER), and ricin toxin A chain (RTA) enters the cytosol from the ER. Previous reports indicated that RTA inhibits activation of the unfolded protein response (UPR) in yeast and in mammalian cells. Both precursor (preRTA) and mature form of RTA (mRTA) inhibited splicing of HAC1u (u for uninduced) mRNA, suggesting that UPR inhibition occurred on the cytosolic face of the ER. Here, we examined the role of ribosome binding and depurination activity on inhibition of the UPR using mRTA mutants. An active-site mutant with very low depurination activity, which bound ribosomes as WT RTA, did not inhibit HAC1u mRNA splicing. A ribosome-binding mutant, which showed reduced binding to ribosomes but retained depurination activity, inhibited HAC1u mRNA splicing. This mutant allowed separation of the UPR inhibition by RTA from cytotoxicity because it reduced the rate of depurination. The ribosome-binding mutant inhibited the UPR without affecting IRE1 oligomerization or cleavage of HAC1u mRNA at the splice site junctions. Inhibition of the UPR correlated with the depurination level, suggesting that ribosomes play a role in splicing of HAC1u mRNA. We show that HAC1u mRNA is associated with ribosomes and does not get processed on depurinated ribosomes, thereby inhibiting the UPR. These results demonstrate that RTA inhibits HAC1u mRNA splicing through its depurination activity on the ribosome without directly affecting IRE1 oligomerization or the splicing reaction and provide evidence that IRE1 recognizes HAC1u mRNA that is associated with ribosomes.
Collapse
Affiliation(s)
- Michael Pierce
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520
| | - Diana Vengsarkar
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520
| | - John E McLaughlin
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520
| | - Jennifer N Kahn
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520
| | - Nilgun E Tumer
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520
| |
Collapse
|
40
|
Dianthin and Its Potential in Targeted Tumor Therapies. Toxins (Basel) 2019; 11:toxins11100592. [PMID: 31614697 PMCID: PMC6832487 DOI: 10.3390/toxins11100592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 12/26/2022] Open
Abstract
Dianthin enzymes belong to ribosome-inactivating proteins (RIPs) of type 1, i.e., they only consist of a catalytic domain and do not have a cell binding moiety. Dianthin-30 is very similar to saporin-S3 and saporin-S6, two RIPs often used to design targeted toxins for tumor therapy and already tested in some clinical trials. Nevertheless, dianthin enzymes also exhibit differences to saporin with regard to structure, efficacy, toxicity, immunogenicity and production by heterologous expression. Some of the distinctions might make dianthin more suitable for targeted tumor therapies than other RIPs. The present review provides an overview of the history of dianthin discovery and illuminates its structure, function and role in targeted toxins. It further discusses the option to increase the efficacy of dianthin by endosomal escape enhancers.
Collapse
|
41
|
Application of therapeutic protein-based fusion toxins. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0040-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
42
|
Bich Loan NT, Trung NN, Le Na NT, Thang ND. Anticancer Activities of Ricin-Liposome Complexes on SKMEL-28 Cells. Asian Pac J Cancer Prev 2019; 20:2117-2123. [PMID: 31350974 PMCID: PMC6745201 DOI: 10.31557/apjcp.2019.20.7.2117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Indexed: 01/03/2023] Open
Abstract
Background: Ricin has been reported as a potential chemical for cancer treatment. However, so far, the application
of ricin in cancer treatment is very limited because of its non-specificity. Methods: In this study, ricin were conjugated/
encapsulated with DOTAP/DOPE liposome to form ricin-liposome complexes (ricin-lipososme1, ricin-liposome2,
ricin-liposome3 and ricin-liposome4). Characteristics of ricin-liposome complexes were analyzed and their effects on
survival, apoptosis, migration, invasion and tumor formation of SKMEL-28 melanoma cells were examined by carrying
out the MTT assay, apoptosis assay, scratch wound healing assay, invasion assay and soft-agar colony formation assay,
respectively. Results: Ricin-liposome complexes had even size-distribution with average size of around 340 nm. These
ricin-liposome complexes were able to penetrate into the cells via endocytosis with the highest ability of the ricinliposome3.
It also showed that ricin-liposome3 expressed very high toxicity with the IC50 of 62.4 ng/mL and followed
by ricin-liposome4 (286.4 mg/mL), ricin-liposome2 (417.5 ng/mL), and ricin-liposome1 (604.3 ng/mL) to SKMEL-28
cells at 36 hours post treatment. At the concentrations of IC10 (10.1 ng/mL), ricin-liposome3 strongly induced necrosis
and apoptosis of SKMEL-28 cells up to 25.6% and 11.4%, respectively. Moreover, ricin-liposome3 expressed great
anticancer properties by decreasing the migration, invasion and tumor formation abilities of SKMEL-28 cells of 7.5
folds, 4.3 folds and 5.9 folds, respectively, compared with those of control SKMEL-28 cells. Conclusion: The obtained
results from our study suggest that although ricin is listed as one of the most poisonous substances in nature, it can be
used in the complex forms with liposome to increase its specificity to apply in treatment of melanoma and other cancers.
Collapse
Affiliation(s)
- Nguyen Thi Bich Loan
- Department of Biochemistry and Molecular Biology, VNU University of Science, Vietnam National University, Vietnam. ,Unit of Therapeutic Chemistry and Pharmacognosy, University of Mons (UMONS), Belgium
| | - Ngo Ngoc Trung
- Institute of Chemistry and Environment, High Command of Chemistry, Ministry of National Defense, Vietnam
| | - Nguyen Thi Le Na
- Department of Biochemistry and Molecular Biology, VNU University of Science, Vietnam National University, Vietnam.
| | - Nguyen Dinh Thang
- Department of Biochemistry and Molecular Biology, VNU University of Science, Vietnam National University, Vietnam. ,The Key Laboratory of Enzyme and Protein Technology, VNU University of Science, Vietnam National University, Vietnam
| |
Collapse
|
43
|
Intracellular Transport and Cytotoxicity of the Protein Toxin Ricin. Toxins (Basel) 2019; 11:toxins11060350. [PMID: 31216687 PMCID: PMC6628406 DOI: 10.3390/toxins11060350] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
Ricin can be isolated from the seeds of the castor bean plant (Ricinus communis). It belongs to the ribosome-inactivating protein (RIP) family of toxins classified as a bio-threat agent due to its high toxicity, stability and availability. Ricin is a typical A-B toxin consisting of a single enzymatic A subunit (RTA) and a binding B subunit (RTB) joined by a single disulfide bond. RTA possesses an RNA N-glycosidase activity; it cleaves ribosomal RNA leading to the inhibition of protein synthesis. However, the mechanism of ricin-mediated cell death is quite complex, as a growing number of studies demonstrate that the inhibition of protein synthesis is not always correlated with long term ricin toxicity. To exert its cytotoxic effect, ricin A-chain has to be transported to the cytosol of the host cell. This translocation is preceded by endocytic uptake of the toxin and retrograde traffic through the trans-Golgi network (TGN) and the endoplasmic reticulum (ER). In this article, we describe intracellular trafficking of ricin with particular emphasis on host cell factors that facilitate this transport and contribute to ricin cytotoxicity in mammalian and yeast cells. The current understanding of the mechanisms of ricin-mediated cell death is discussed as well. We also comment on recent reports presenting medical applications for ricin and progress associated with the development of vaccines against this toxin.
Collapse
|
44
|
Ricin: An Ancient Story for a Timeless Plant Toxin. Toxins (Basel) 2019; 11:toxins11060324. [PMID: 31174319 PMCID: PMC6628454 DOI: 10.3390/toxins11060324] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022] Open
Abstract
The castor plant (Ricinus communis L.) has been known since time immemorial in traditional medicine in the pharmacopeia of Mediterranean and eastern ancient cultures. Moreover, it is still used in folk medicine worldwide. Castor bean has been mainly recommended as anti-inflammatory, anthelmintic, anti-bacterial, laxative, abortifacient, for wounds, ulcers, and many other indications. Many cases of human intoxication occurred accidentally or voluntarily with the ingestion of castor seeds or derivatives. Ricinus toxicity depends on several molecules, among them the most important is ricin, a protein belonging to the family of ribosome-inactivating proteins. Ricin is the most studied of this category of proteins and it is also known to the general public, having been used for several biocrimes. This manuscript intends to give the reader an overview of ricin, focusing on the historical path to the current knowledge on this protein. The main steps of ricin research are here reported, with particular regard to its enzymatic activity, structure, and cytotoxicity. Moreover, we discuss ricin toxicity for animals and humans, as well as the relation between bioterrorism and ricin and its impact on environmental toxicity. Ricin has also been used to develop immunotoxins for the elimination of unwanted cells, mainly cancer cells; some of these immunoconjugates gave promising results in clinical trials but also showed critical limitation.
Collapse
|
45
|
Zhu Y, Weldon JE. Evaluating the influence of common antibiotics on the efficacy of a recombinant immunotoxin in tissue culture. BMC Res Notes 2019; 12:293. [PMID: 31133049 PMCID: PMC6537151 DOI: 10.1186/s13104-019-4337-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/22/2019] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE Recombinant immunotoxins (RITs) are antibody-toxin fusion proteins that can selectively eliminate populations of cells expressing specific surface receptors. They are in evaluation as therapeutic agents for cancer. RITs based on Pseudomonas exotoxin A (PE) are in use clinically for the treatment of hairy cell leukemia, and under trial for the treatment of other cancers. In an effort to improve the efficacy of PE-based RITs, we evaluated the potential of combination therapy with several common antibiotics (tetracycline, chloramphenicol, streptomycin, linezolid, fusidic acid, and kanamycin) on human cell lines HEK293, OVCAR8, and CA46. Antibiotics were selected based on their potential to inhibit mitochondrial protein synthesis and disrupt energy metabolism in cancer cells. RESULTS Tetracycline, chloramphenicol, linezolid, and fusidic acid alone killed cultured human cells at high concentrations. At high but nontoxic concentrations of each antibiotic, only chloramphenicol treatment of the Burkitt's lymphoma cell line CA46 showed enhanced cytotoxicity when paired with an anti-transferrin receptor/PE RIT. This result, however, could not be replicated in additional Burkitt's lymphoma cell lines Ramos and Raji. Although the six antibiotics we tested are not promising candidates for RIT combination therapy, we suggest that fusidic acid could be considered independently as a potential cancer therapeutic.
Collapse
Affiliation(s)
- Yuyi Zhu
- Department of Biological Sciences, The Jess and Mildred Fisher College of Science and Mathematics, Towson University, Towson, MD 21252 USA
| | - John E. Weldon
- Department of Biological Sciences, The Jess and Mildred Fisher College of Science and Mathematics, Towson University, Towson, MD 21252 USA
| |
Collapse
|
46
|
Bortolotti M, Mercatelli D, Polito L. Momordica charantia, a Nutraceutical Approach for Inflammatory Related Diseases. Front Pharmacol 2019; 10:486. [PMID: 31139079 PMCID: PMC6517695 DOI: 10.3389/fphar.2019.00486] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/17/2019] [Indexed: 01/24/2023] Open
Abstract
Momordica charantia, commonly called bitter melon, is a plant belonging to Cucurbitaceae family known for centuries for its pharmacological activities, and nutritional properties. Due to the presence of many bioactive compounds, some of which possess potent biological actions, this plant is used in folk medicine all over the world for the treatment of different pathologies, mainly diabetes, but also cancer, and other inflammation-associated diseases. It is widely demonstrated that M. charantia extracts contribute in lowering glycaemia in patients affected by type 2 diabetes. However, the majority of existing studies on M. charantia bioactive compounds were performed only on cell lines and in animal models. Therefore, because the real impact of bitter melon on human health has not been thoroughly demonstrated, systematic clinical studies are needed to establish its efficacy and safety in patients. Besides, both in vitro and in vivo studies have demonstrated that bitter melon may also elicit toxic or adverse effects under different conditions. The aim of this review is to provide an overview of anti-inflammatory and anti-neoplastic properties of bitter melon, discussing its pharmacological activity as well as the potential adverse effects. Even if a lot of literature is available about bitter melon as antidiabetic drug, few papers discuss the anti-inflammatory and anti-cancer properties of this plant.
Collapse
Affiliation(s)
- Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Daniele Mercatelli
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
47
|
Barkhordari F, Sohrabi N, Davami F, Mahboudi F, Garoosi YT. Cloning, expression and characterization of a HER2-alpha luffin fusion protein in Escherichia coli. Prep Biochem Biotechnol 2019; 49:759-766. [PMID: 31032734 DOI: 10.1080/10826068.2019.1608447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In recent decades, immunotoxins have attracted significant attention in treatment of a wide range of diseases including cancers due to their natural origins and their role in blocking crucial pathways within the cells. Ribosome inactivating proteins (RIPs) are efficient molecules in blocking protein synthesis through interactions with ribosomal rRNA molecules. cDNA molecule encoding HER2 scFv antibody fragment originated from trastuzumab attached to the mature alpha luffin gene fragment was subcloned into pET28a expression vector and expressed in different E. coli expression hosts. Identity of the expressed recombinant protein was investigated through western blotting and the fusion protein was purified using Ni-NTA affinity chromatography. The biological activity (toxicity) of the protein was investigated on DNA and RNA samples. A 58 kDa protein was expressed in E. coli. The best protein expression level was achieved in 0.2 mM IPTG at 30 °C in TB medium using E. coli BL21 (DE3) host strain. The fusion protein showed RNase and DNA glycosylase activity on tested RNA and DNA samples. DNA glycosylase activity of the recombinant fusion protein showed that alpha luffin part of this protein is active in conjugation to the scFv molecule and the expressed protein can be further studied in targeted biological in vitro assays.
Collapse
Affiliation(s)
- Farzaneh Barkhordari
- a Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran.,b Department of Biochemistry, Payame Noor University , Tehran , Iran
| | - Nooshin Sohrabi
- c Department of Biology, Payame Noor University , Tehran , Iran
| | - Fatemeh Davami
- a Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | - Fereidoun Mahboudi
- a Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | | |
Collapse
|
48
|
Kokorin A, Weise C, Sama S, Weng A. A new type 1 ribosome-inactivating protein from the seeds of Gypsophila elegans M.Bieb. PHYTOCHEMISTRY 2019; 157:121-127. [PMID: 30399494 DOI: 10.1016/j.phytochem.2018.10.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/16/2018] [Accepted: 10/25/2018] [Indexed: 06/08/2023]
Abstract
Ribosome-inactivating proteins (RIPs) are enzymes with N-glycosylase activity that remove adenine bases from the ribosomal RNA. In theory, one single RIP molecule internalized into a cell is sufficient to induce cell death. For this reason, RIPs are of high potential as toxic payload for anti-tumor therapy. A considerable number of RIPs are synthesized by plants that belong to the carnation family (Caryophyllaceae). Prominent examples are the RIPs saporin from Saponaria officinalis L. or dianthin from Dianthus caryophyllus L. In this study, we have isolated and characterized a novel RIP (termed gypsophilin-S) from the tiny seeds of Gypsophila elegans M. Bieb. (Caryophyllaceae). It is noteworthy that this is the first study presenting the complete amino acid sequence of a RIP from a Gypsophila species. Gypsophilin-S was isolated from the defatted seed material following ammonium sulphate precipitation and HPLC-based ion exchange chromatography. Gypsophilin-S-containing fractions were analysed by SDS-PAGE and mass spectrometry. The full amino acid sequence of gypsophilin-S was assembled by MALDI-TOF-MS-MS and PCR. Gypsophilin-S exhibited strong adenine releasing activity and its cytotoxicity in human glioblastoma cells was investigated using an impedance-based real-time assay in comparison with recombinant saporin and dianthin.
Collapse
Affiliation(s)
- Arsenij Kokorin
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Christoph Weise
- Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Simko Sama
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Alexander Weng
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany.
| |
Collapse
|
49
|
Ovacik M, Lin K. Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development. Clin Transl Sci 2018; 11:540-552. [PMID: 29877608 PMCID: PMC6226118 DOI: 10.1111/cts.12567] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
The tutorial introduces the readers to the fundamentals of antibody pharmacokinetics (PK) in the context of drug development. Topics covered include an overview of antibody development, PK characteristics, and the application of antibody PK/pharmacodynamics (PD) in research and development decision-making. We also discuss the general considerations for planning a nonclinical PK program and describe the types of PK studies that should be performed during early development of monoclonal antibodies.
Collapse
Affiliation(s)
- Meric Ovacik
- Department of Preclinical and Translational Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Kedan Lin
- Clinical Pharmacology, NGM Biopharmaceuticals, Inc., South San Francisco, California, USA
| |
Collapse
|
50
|
Díaz R, Pallarès V, Cano-Garrido O, Serna N, Sánchez-García L, Falgàs A, Pesarrodona M, Unzueta U, Sánchez-Chardi A, Sánchez JM, Casanova I, Vázquez E, Mangues R, Villaverde A. Selective CXCR4 + Cancer Cell Targeting and Potent Antineoplastic Effect by a Nanostructured Version of Recombinant Ricin. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800665. [PMID: 29845742 DOI: 10.1002/smll.201800665] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/24/2018] [Indexed: 05/14/2023]
Abstract
Under the unmet need of efficient tumor-targeting drugs for oncology, a recombinant version of the plant toxin ricin (the modular protein T22-mRTA-H6) is engineered to self-assemble as protein-only, CXCR4-targeted nanoparticles. The soluble version of the construct self-organizes as regular 11 nm planar entities that are highly cytotoxic in cultured CXCR4+ cancer cells upon short time exposure, with a determined IC50 in the nanomolar order of magnitude. The chemical inhibition of CXCR4 binding sites in exposed cells results in a dramatic reduction of the cytotoxic potency, proving the receptor-dependent mechanism of cytotoxicity. The insoluble version of T22-mRTA-H6 is, contrarily, moderately active, indicating that free, nanostructured protein is the optimal drug form. In animal models of acute myeloid leukemia, T22-mRTA-H6 nanoparticles show an impressive and highly selective therapeutic effect, dramatically reducing the leukemia cells affectation of clinically relevant organs. Functionalized T22-mRTA-H6 nanoparticles are then promising prototypes of chemically homogeneous, highly potent antitumor nanostructured toxins for precise oncotherapies based on self-mediated intracellular drug delivery.
Collapse
Affiliation(s)
- Raquel Díaz
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Victor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Olivia Cano-Garrido
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Laura Sánchez-García
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Aïda Falgàs
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Mireia Pesarrodona
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | | | - Julieta M Sánchez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT) (CONICET-Universidad Nacional de Córdoba), ICTA and Cátedra de Química Biológica, Departamento de Química, FCEFyN, UNC, Av. Velez Sarsfield 1611, X 5016GCA, Córdoba, Argentina
| | - Isolda Casanova
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| |
Collapse
|