1
|
Yu J, An N, Zhu J, Zhu B, Zhang G, Chen K, Zhou Y, Ye T, Li G. AVL-armed oncolytic vaccinia virus promotes viral replication and boosts antitumor immunity via increasing ROS levels in pancreatic cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200878. [PMID: 39431173 PMCID: PMC11488421 DOI: 10.1016/j.omton.2024.200878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 10/22/2024]
Abstract
Pancreatic malignant neoplasm is an extremely deadly malignancy well known for its resistance to traditional therapeutic approaches. Enhanced treatments are imperative for individuals diagnosed with pancreatic cancer (PC). Recent investigations have shed light on the wide-ranging anticancer properties of genetic therapy facilitated by oncolytic vaccinia virus. To illuminate the precise impacts of Aphrocallistes vastus lectin-armed oncolytic vaccinia virus (oncoVV-AVL) on PC, AsPC-1 and PANC-1 cells underwent treatment with oncoVV-AVL. Our findings revealed that oncoVV-AVL possesses the capacity to heighten oncolytic effects on PC cells and incite the production of diverse cytokines like tumor necrosis factor-α, interleukin-6 (IL-6), IL-8, and interferon-I (IFN-I), without triggering antiviral responses. Additionally, oncoVV-AVL can significantly elevate the levels of ROS in PC cells, initiating an oxidative stress response that promotes viral replication, apoptosis, and autophagy. Moreover, in xenograft tumor models, oncoVV-AVL notably restrained PC growth, enhanced IFN-γ levels in the bloodstream, and reprogrammed macrophages. Our investigation indicates that oncoVV-AVL boosts the efficacy of antitumor actions against PC tumors by orchestrating reactive oxygen species-triggered viral replication, fostering M1 polarization, and reshaping the tumor microenvironment to transform cold PC tumors into hot ones. These findings imply that oncoVV-AVL could present a novel therapeutic approach for treating PC tumors.
Collapse
Affiliation(s)
- Jianlei Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Nan An
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jili Zhu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Borong Zhu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Guohui Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Kan Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yanrong Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Ting Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Gongchu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
2
|
Buriak I, Kumeiko V. Marine Lectins and Lectin-like Proteins as Promising Molecules Targeting Aberrant Glycosylation Signatures in Human Brain Tumors. Mar Drugs 2024; 22:527. [PMID: 39728102 DOI: 10.3390/md22120527] [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: 10/20/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Glycosylation is a ubiquitous and the most structurally diverse post-translational modification of proteins. High levels of phenotypic heterogeneity in brain tumors affect the biosynthetic pathway of glycosylation machinery, resulting in aberrant glycosylation patterns. Traditionally, unique glycocode readers, carbohydrate-binding proteins, have been used to identify differentially expressed carbohydrate determinants associated with the tumor cell surface. However, identifying novel distinctive glycosylation signatures in brain tumors requires the timely development of molecular tools capable of targeting them. We classified marine-derived lectins and lectin-like molecules according to their ability to cover aberrant glycosylation patterns in brain tumors to encourage exploration of the potential of these molecules for precision diagnostics and personalized therapy.
Collapse
Affiliation(s)
- Ivan Buriak
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| | - Vadim Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| |
Collapse
|
3
|
Bala Subramaniyan S, Veerappan A. Lectins as the prominent potential to deliver bioactive metal nanoparticles by recognizing cell surface glycans. Heliyon 2024; 10:e29394. [PMID: 38638961 PMCID: PMC11024627 DOI: 10.1016/j.heliyon.2024.e29394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Lectins are renowned for recognizing specific carbohydrates, but there is evidence that they can bind to other endogenous ligands. Therefore, lectin can be used as a carrier to recognize glycoconjugates on the cell surface. The anticancer, antibacterial, and immunomodulatory properties of some lectins are established. Metal nanoparticles (MNPs) have been used in various fields recently, but their documented toxicity has raised questions about their suitability for biomedical uses. The advantages of MNPs can be realized if we deliver the NPs to the site of action; as a result, NPs may achieve greater therapeutic efficiency at lower doses with less toxicity. The use of carbohydrate specificity by lectin MNPs conjugates for diagnostics and therapeutics was addressed. The review summarised the multidimensional application of lectins and described their potential for delivery of MNPs in future drug development.
Collapse
Affiliation(s)
- Siva Bala Subramaniyan
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, 613 401, Tamil Nadu, India
| | - Anbazhagan Veerappan
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, 613 401, Tamil Nadu, India
| |
Collapse
|
4
|
Grinchenko A, Buriak I, Kumeiko V. Invertebrate C1q Domain-Containing Proteins: Molecular Structure, Functional Properties and Biomedical Potential. Mar Drugs 2023; 21:570. [PMID: 37999394 PMCID: PMC10672478 DOI: 10.3390/md21110570] [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: 09/02/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/25/2023] Open
Abstract
C1q domain-containing proteins (C1qDC proteins) unexpectedly turned out to be widespread molecules among a variety of invertebrates, despite their lack of an integral complement system. Despite the wide distribution in the genomes of various invertebrates, data on the structure and properties of the isolated and characterized C1qDC proteins, which belong to the C1q/TNF superfamily, are sporadic, although they hold great practical potential for the creation of new biotechnologies. This review not only summarizes the current data on the properties of already-isolated or bioengineered C1qDC proteins but also projects further strategies for their study and biomedical application. It has been shown that further broad study of the carbohydrate specificity of the proteins can provide great opportunities, since for many of them only interactions with pathogen-associated molecular patterns (PAMPs) was evaluated and their antimicrobial, antiviral, and fungicidal activities were studied. However, data on the properties of C1qDC proteins, which researchers originally discovered as lectins and therefore studied their fine carbohydrate specificity and antitumor activity, intriguingly show the great potential of this family of proteins for the creation of targeted drug delivery systems, vaccines, and clinical assays for the differential diagnosis of cancer. The ability of invertebrate C1qDC proteins to recognize patterns of aberrant glycosylation of human cell surfaces and interact with mammalian immunoglobulins indicates the great biomedical potential of these molecules.
Collapse
Affiliation(s)
- Andrei Grinchenko
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.G.); (I.B.)
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| | - Ivan Buriak
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.G.); (I.B.)
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| | - Vadim Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.G.); (I.B.)
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| |
Collapse
|
5
|
Rao MRP, Ghadge I, Kulkarni S, R. Madgulkar A. Importance of Plant Secondary Metabolites in Modern Therapy. REFERENCE SERIES IN PHYTOCHEMISTRY 2023:1-31. [DOI: 10.1007/978-3-031-30037-0_5-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 01/05/2025]
|
6
|
Padmanaban D, Samuel A, Sahayanathan GJ, Raja K, Chinnasamy A. Anticancer effect of marine bivalves derived polysaccharides against human cancer cells. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2022. [DOI: 10.1016/j.bcab.2021.102240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Grinchenko AV, von Kriegsheim A, Shved NA, Egorova AE, Ilyaskina DV, Karp TD, Goncharov NV, Petrova IY, Kumeiko VV. A Novel C1q Domain-Containing Protein Isolated from the Mollusk Modiolus kurilensis Recognizing Glycans Enriched with Acidic Galactans and Mannans. Mar Drugs 2021; 19:668. [PMID: 34940667 PMCID: PMC8706970 DOI: 10.3390/md19120668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/18/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
C1q domain-containing (C1qDC) proteins are a group of biopolymers involved in immune response as pattern recognition receptors (PRRs) in a lectin-like manner. A new protein MkC1qDC from the hemolymph plasma of Modiolus kurilensis bivalve mollusk widespread in the Northwest Pacific was purified. The isolation procedure included ammonium sulfate precipitation followed by affinity chromatography on pectin-Sepharose. The full-length MkC1qDC sequence was assembled using de novo mass-spectrometry peptide sequencing complemented with N-terminal Edman's degradation, and included 176 amino acid residues with molecular mass of 19 kDa displaying high homology to bivalve C1qDC proteins. MkC1qDC demonstrated antibacterial properties against Gram-negative and Gram-positive strains. MkC1qDC binds to a number of saccharides in Ca2+-dependent manner which characterized by structural meta-similarity in acidic group enrichment of galactose and mannose derivatives incorporated in diversified molecular species of glycans. Alginate, κ-carrageenan, fucoidan, and pectin were found to be highly effective inhibitors of MkC1qDC activity. Yeast mannan, lipopolysaccharide (LPS), peptidoglycan (PGN) and mucin showed an inhibitory effect at concentrations three orders of magnitude greater than for the most effective saccharides. MkC1qDC localized to the mussel hemal system and interstitial compartment. Intriguingly, MkC1qDC was found to suppress proliferation of human adenocarcinoma HeLa cells in a dose-dependent manner, indicating to the biomedical potential of MkC1qDC protein.
Collapse
Affiliation(s)
- Andrei V. Grinchenko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (A.V.G.); (N.A.S.); (N.V.G.); (I.Y.P.)
| | - Alex von Kriegsheim
- Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK;
| | - Nikita A. Shved
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (A.V.G.); (N.A.S.); (N.V.G.); (I.Y.P.)
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.E.E.); (D.V.I.); (T.D.K.)
| | - Anna E. Egorova
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.E.E.); (D.V.I.); (T.D.K.)
| | - Diana V. Ilyaskina
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.E.E.); (D.V.I.); (T.D.K.)
| | - Tatiana D. Karp
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.E.E.); (D.V.I.); (T.D.K.)
| | - Nikolay V. Goncharov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (A.V.G.); (N.A.S.); (N.V.G.); (I.Y.P.)
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.E.E.); (D.V.I.); (T.D.K.)
| | - Irina Y. Petrova
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (A.V.G.); (N.A.S.); (N.V.G.); (I.Y.P.)
| | - Vadim V. Kumeiko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (A.V.G.); (N.A.S.); (N.V.G.); (I.Y.P.)
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.E.E.); (D.V.I.); (T.D.K.)
| |
Collapse
|
8
|
Antitumor Potential of Marine and Freshwater Lectins. Mar Drugs 2019; 18:md18010011. [PMID: 31877692 PMCID: PMC7024344 DOI: 10.3390/md18010011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Often, even the most effective antineoplastic drugs currently used in clinic do not efficiently allow complete healing due to the related toxicity. The reason for the toxicity lies in the lack of selectivity for cancer cells of the vast majority of anticancer agents. Thus, the need for new potent anticancer compounds characterized by a better toxicological profile is compelling. Lectins belong to a particular class of non-immunogenic glycoproteins and have the characteristics to selectively bind specific sugar sequences on the surface of cells. This property is exploited to exclusively bind cancer cells and exert antitumor activity through the induction of different forms of regulated cell death and the inhibition of cancer cell proliferation. Thanks to the extraordinary biodiversity, marine environments represent a unique source of active natural compounds with anticancer potential. Several marine and freshwater organisms, ranging from the simplest alga to the most complex vertebrate, are amazingly enriched in these proteins. Remarkably, all studies gathered in this review show the impressive anticancer effect of each studied marine lectin combined with irrelevant toxicity in vitro and in vivo and pave the way to design clinical trials to assess the real antineoplastic potential of these promising proteins. It provides a concise and precise description of the experimental results, their interpretation as well as the experimental conclusions that can be drawn.
Collapse
|
9
|
Li S, Ruan Z, Yang X, Li M, Yang D. Immune recognition, antimicrobial and opsonic activities mediated by a sialic acid binding lectin from Ruditapes philippinarum. FISH & SHELLFISH IMMUNOLOGY 2019; 93:66-72. [PMID: 31306758 DOI: 10.1016/j.fsi.2019.07.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023]
Abstract
In the present study, a sialic acid-binding lectin was identified and characterized from Manila clam Ruditapes philippinarum (designed as RpSABL-1). Multiple alignments strongly suggested that RpSABL-1 was a new member of the sialic acid-binding lectin family. In non-stimulated clams, RpSABL-1 transcripts were constitutively expressed in all five tested tissues, especially in hepatopancreas. After Vibrio anguillarum challenge, the expression of RpSABL-1 mRNA was significantly up-regulated at 6 h (P < 0.05), 12 h (P < 0.01) and 24 h (P < 0.01). Recombinant RpSABL-1 protein (rRpSABL-1) displayed apparent binding activities towards lipopolysaccharides (LPS) and peptidoglycan (PGN), but not to glucan or chitin in vitro. Coinciding with the PAMPs binding assay, rRpSABL-1 exhibited obvious agglutination activities against Gram-positive bacterium Staphyloccocus aureus, Gram-negative bacteria Escherichia coli, V. anguillarum and Vibrio harveyi. Meanwhile, rRpSABL-1 showed antibacterial activities against E. coli, and biofilm formation of E. coli could also be inhibited after incubated with rRpSABL-1. Moreover, the encapsulation, phagocytosis and chemotactic ability of hemocytes could be enhanced by rRpSABL-1. All these results suggested that RpSABL-1 could function as a pattern recognition receptor with versatile functions in the innate immune responses of R. philippinarum.
Collapse
Affiliation(s)
- Shengqiang Li
- College of Agriculture, Ludong University, Yantai, PR China
| | - Zeli Ruan
- College of Agriculture, Ludong University, Yantai, PR China
| | - Xiyun Yang
- College of Agriculture, Ludong University, Yantai, PR China
| | - Mingzhu Li
- College of Agriculture, Ludong University, Yantai, PR China.
| | - Dinglong Yang
- Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, PR China; Center for Ocean Mega-science, Chinese Academy of Sciences, Qingdao, Shandong, 266071, PR China.
| |
Collapse
|
10
|
Wu T, Xiang Y, Liu T, Wang X, Ren X, Ye T, Li G. Oncolytic Vaccinia Virus Expressing Aphrocallistes vastus Lectin as a Cancer Therapeutic Agent. Mar Drugs 2019; 17:md17060363. [PMID: 31248066 PMCID: PMC6628141 DOI: 10.3390/md17060363] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022] Open
Abstract
Lectins display a variety of biological functions including insecticidal, antimicrobial, as well as antitumor activities. In this report, a gene encoding Aphrocallistes vastus lectin (AVL), a C-type lectin, was inserted into an oncolytic vaccinia virus vector (oncoVV) to form a recombinant virus oncoVV-AVL, which showed significant in vitro antiproliferative activity in a variety of cancer cell lines. Further investigations revealed that oncoVV-AVL replicated faster than oncoVV significantly in cancer cells. Intracellular signaling elements including NF-κB2, NIK, as well as ERK were determined to be altered by oncoVV-AVL. Virus replication upregulated by AVL was completely dependent on ERK activity. Furthermore, in vivo studies showed that oncoVV-AVL elicited significant antitumor effect in colorectal cancer and liver cancer mouse models. Our study might provide insights into a novel way of the utilization of marine lectin AVL in oncolytic viral therapies.
Collapse
Affiliation(s)
- Tao Wu
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Yulin Xiang
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Tingting Liu
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Xue Wang
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Xiaoyuan Ren
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Ting Ye
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Gongchu Li
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
11
|
Sialic Acid-Binding Lectin from Bullfrog Eggs Exhibits an Anti-Tumor Effect Against Breast Cancer Cells Including Triple-Negative Phenotype Cells. Molecules 2018; 23:molecules23102714. [PMID: 30347895 PMCID: PMC6222625 DOI: 10.3390/molecules23102714] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/19/2018] [Accepted: 10/19/2018] [Indexed: 01/22/2023] Open
Abstract
Sialic acid-binding lectin from Rana catesbeiana eggs (cSBL) is a multifunctional protein that has lectin and ribonuclease activity. In this study, the anti-tumor activities of cSBL were assessed using a panel of breast cancer cell lines. cSBL suppressed the cell growth of all cancer cell lines tested here at a concentration that is less toxic, or not toxic at all, to normal cells. The growth suppressive effect was attributed to the cancer-selective induction of apoptosis. We assessed the expressions of several key molecules associated with the breast cancer phenotype after cSBL treatment by western blotting. cSBL decreased the expression level of estrogen receptor (ER) α, while it increased the phosphorylation level of p38 mitogen-activated protein kinase (MAPK). cSBL also suppressed the expression of the progesterone receptor (PgR) and human epidermal growth factor receptor type 2 (HER2). Furthermore, it was revealed that cSBL decreases the expression of the epidermal growth factor receptor (EGFR/HER1) in triple-negative breast cancer cells. These results indicate that cSBL induces apoptosis with decreasing ErbB family proteins and may have great potential for breast cancer chemotherapy, particularly in triple-negative phenotype cells.
Collapse
|
12
|
Li G, Mei S, Cheng J, Wu T, Luo J. Haliotis discus discus Sialic Acid-Binding Lectin Reduces the Oncolytic Vaccinia Virus Induced Toxicity in a Glioblastoma Mouse Model. Mar Drugs 2018; 16:md16050141. [PMID: 29701680 PMCID: PMC5983273 DOI: 10.3390/md16050141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/22/2018] [Accepted: 04/25/2018] [Indexed: 12/25/2022] Open
Abstract
Although oncolytic viruses provide attractive vehicles for cancer treatment, their adverse effects are largely ignored. In this work, rat C6 glioblastoma cells were subcutaneously xenografted into mice, and a thymidine kinase-deficient oncolytic vaccinia virus (oncoVV) induced severe toxicity in this model. However, oncoVV-HddSBL, in which a gene encoding Haliotis discus discus sialic acid-binding lectin (HddSBL) was inserted into oncoVV, significantly prolonged the survival of mice as compared to the control virus. HddSBL reduced the tumor secreted serum rat IL-2 level upregulated by oncoVV, promoted viral replication, as well as inhibited the expression of antiviral factors in C6 glioblastoma cell line. Furthermore, HddSBL downregulated the expression levels of histone H3 and H4, and upregulated histone H3R8 and H4R3 asymmetric dimethylation, confirming the effect of HddSBL on chromatin structure suggested by the transcriptome data. Our results might provide insights into the utilization of HddSBL in counteracting the adverse effects of oncolytic vaccinia virus.
Collapse
Affiliation(s)
- Gongchu Li
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Shengsheng Mei
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Jianhong Cheng
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Tao Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Jingjing Luo
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
13
|
Tatsuta T, Satoh T, Sugawara S, Hara A, Hosono M. Sialic acid-binding lectin from bullfrog eggs inhibits human malignant mesothelioma cell growth in vitro and in vivo. PLoS One 2018; 13:e0190653. [PMID: 29298350 PMCID: PMC5752036 DOI: 10.1371/journal.pone.0190653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/18/2017] [Indexed: 12/29/2022] Open
Abstract
Malignant mesothelioma is an aggressive cancer that results from exposure to asbestos. The therapeutic options for this type of cancer are limited; therefore, the development of novel therapeutic agents is urgently required. Sialic acid-binding lectin isolated from Rana catesbeiana oocytes (cSBL) is a novel therapeutic candidate for cancer, which exhibits antitumor activity mediated through RNA degradation. In the present study, we evaluated the effect of cSBL in vitro and in vivo. Xenograft-competent H2452 and MSTO human mesothelioma cell lines were treated with cSBL, and the pathway by which cSBL induces apoptosis was analyzed. In vivo studies were performed using nude mice inoculated with one of the two cell lines, and the effects of cSBL and pemetrexed were monitored simultaneously. Furthermore, the pharmacological interactions between the three agents (pemetrexed, cisplatin and cSBL) were statistically assessed. It was demonstrated that cSBL treatments caused morphological and biochemical apoptotic changes in both cell lines. Caspase cascade analysis revealed that an intrinsic pathway mediated cSBL-induced apoptosis. The administration of cSBL significantly inhibited tumor growth in two xenograft models, without any adverse effects. Furthermore, the combination index and dose reduction index values indicated that the cSBL + pemetrexed combination showed the highest synergism, and thus potential for reducing dosage of each drug, compared with the other combinations, including the existing pemetrexed + cisplatin regimen. cSBL exerted prominent antitumor effects on malignant mesothelioma cells in vitro and in vivo, and showed favorable effects when combined with pemetrexed. These results suggest that cSBL has potential as a novel drug for the treatment of malignant mesothelioma.
Collapse
Affiliation(s)
- Takeo Tatsuta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Japan
| | - Toshiyuki Satoh
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Japan
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Japan
| | - Shigeki Sugawara
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Japan
| | - Akiyoshi Hara
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Japan
| | - Masahiro Hosono
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Japan
- * E-mail:
| |
Collapse
|
14
|
Li G, Zhao Z, Wu B, Su Q, Wu L, Yang X, Chen J. Ulva pertusa lectin 1 delivery through adenovirus vector affects multiple signaling pathways in cancer cells. Glycoconj J 2017; 34:489-498. [PMID: 28349379 DOI: 10.1007/s10719-017-9767-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 02/26/2017] [Accepted: 03/20/2017] [Indexed: 12/13/2022]
Abstract
Ulva pertusa lectin 1 (UPL1) is a N-acetyl-D-glucosamine (GlcNAc) binding lectin in marine green alga Ulva pertusa. Exogenous UPL1 colocalized with protein arginine methyltransferase 5 (PRMT5), methylosome protein 50 (MEP50), β-actin and β-tubulin, indicating the interaction of UPL1 with the methylosome and cytoskeleton. UPL1 delivery through adenovirus vector (Ad-UPL1) dramatically induced extracellularly regulated protein kinases 1/2 (ERK1/2) phosphorylation in liver cancer cell lines BEL-7404 and Huh7. Signaling pathways including p38 mitogen-activated protein kinase (MAPK), and Akt were also affected by Ad-UPL1 in a cell type dependent manner. MEK1/2 inhibitor U0126, as well as to a lesser extent p38 MAPK inhibitor SB203580 and phosphoinositide 3-kinase (PI3K) inhibitor LY294002, completely eliminated a higher molecular weight isoform of β-tubulin induced by Ad-UPL1, and significantly enhanced the cytotoxicity of Ad-UPL1 in Huh7 cells, suggesting that the inhibition of MEK1/2, p38 MAPK, and PI3K enhanced antiproliferative effect of Ad-UPL1 possibly through regulating the modification of β-tubulin. Ad-UPL1 completely inhibited the expression of autophagy-related factor Beclin1, but induced LC3-II expression in Huh7 cells. In addition, Ad-UPL1 significantly enhanced starvation induced survival suppression in Huh7 cells. Our data elucidated intracellular signaling pathways affected by exogenous UPL1, and may provide insights into a novel way of UPL1 delivery through adenovirus vectors combined with survival signaling inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Gongchu Li
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China.
| | - Zhenzhen Zhao
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Bingbing Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Qunshu Su
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Liqin Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Xinyan Yang
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Jing Chen
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Marine Lectins DlFBL and HddSBL Fused with Soluble Coxsackie-Adenovirus Receptor Facilitate Adenovirus Infection in Cancer Cells BUT Have Different Effects on Cell Survival. Mar Drugs 2017; 15:md15030073. [PMID: 28335432 PMCID: PMC5367030 DOI: 10.3390/md15030073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/26/2017] [Accepted: 03/10/2017] [Indexed: 01/24/2023] Open
Abstract
Cancer development and progression are usually associated with glycosylation change, providing prognostic and diagnostic biomarkers, as well as therapeutic targets, for various cancers. In this work, Dicentrarchus labrax fucose binding lectin (DlFBL) and Haliotis discus discus sialic acid binding lectin (HddSBL) were genetically fused with soluble coxsackie-adenovirus receptor (sCAR), and produced through a bacterial expression system. Results showed that recombinant sCAR-DlFBL not only facilitated adenovirus Ad-EGFP infection in K562/ADR and U87MG cells, but also enhanced the cytotoxicity of adenovirus harboring gene encoding Pinellia pedatisecta agglutinin (PPA) or DlFBL (Ad-PPA or Ad-DlFBL) on U87MG cells through inducing apoptosis. Recombinant sCAR-HddSBL facilitated Ad-EGFP infection, but dramatically counteracted the cytotoxicity of both Ad-PPA and Ad-DlFBL in U87MG cells. Further analysis revealed that sCAR-HddSBL, but not sCAR-DlFBL, significantly upregulated transcription factor E2F1 levels in U87MG cells, which might be responsible for the adverse effect of sCAR-HddSBL on Ad-PPA and Ad-DlFBL. Taken together, our data suggested that sCAR-DlFBL could be further developed to redirect therapeutic adenoviruses to infect cancer cells such as U87MG, and the sCAR-lectin fusion proteins for adenoviral retargeting should be carefully examined for possible survival signaling induced by lectins, such as HddSBL.
Collapse
|
16
|
Kieber-Emmons T, Monzavi-Karbassi B, Hutchins LF, Pennisi A, Makhoul I. Harnessing benefit from targeting tumor associated carbohydrate antigens. Hum Vaccin Immunother 2017; 13:323-331. [PMID: 27929800 PMCID: PMC5328237 DOI: 10.1080/21645515.2017.1264789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Integrating additive or synergistic antitumor effects that focus on distinct elements of tumor biology are the most rational strategies for cancer treatment. Treatments for breast cancer have increased overall survival, but remain limited by lack of efficacy in a subset of breast cancer patients. The real challenge is to define what elements of tumor biology make the most sense to be integrated. An emerging strategy is to consider a systems biology approach to impact multiple interactions in networks as compare with hitting a specific protein-protein interaction target. In this review, we consider how targeting tumor associated carbohydrate antigens (TACA) that are fundamental to signal pathways might be tailored to harness benefit from combination therapy of sustained immunity with chemotherapy. An approach we are developing makes use of a carbohydrate mimetic peptide (CMP) to induce polyspecific antibodies, which by their nature have numerous on and off target effects. Linking multi-target TACA recognition with mechanisms affecting tumor growth in the context of network heterogeneity and concepts of immune surveillance to tumor cells and the type of breast cancer patients that would benefit from such an approach provides a novel integrative treatment.
Collapse
Affiliation(s)
- Thomas Kieber-Emmons
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Laura F. Hutchins
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angela Pennisi
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Issam Makhoul
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
17
|
Chatterjee B, Ghosh K, Yadav N, Kanade SR. A novel L-fucose-binding lectin from Fenneropenaeus indicus induced cytotoxicity in breast cancer cells. J Biochem 2016; 161:87-97. [PMID: 27742744 DOI: 10.1093/jb/mvw057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/20/2016] [Indexed: 01/21/2023] Open
Abstract
Lectins are omnipresent in almost all life forms, being the proteins which specifically bind to carbohydrate moieties on the cell surface; they have been explored for their anti-tumour activities. In this study, we purified a fucose specific-lectin (IFL) from Fenneropenaeus indicus haemolymph using fucose-affinity column and characterized for its haemagglutination activity, carbohydrate specificity, dependency on cations and cytotoxicity against cancer cells. The lectin showed non-specificity against human erythrocytes. It was a Ca2+-dependent lectin which remained stable over wide pH and temperature ranges. The lectin showed effective dose dependent cytotoxicity against different human cancer cell lines and induced apoptosis in MCF-7 cells as evidenced by DNA ladder assay and PARP cleavage in a dose dependent manner. Moreover, an increased p21 level corresponding to cyclin D downregulation in response to IFL treatment was observed which might work as probable factors to inhibit cell growth and induce apoptosis of MCF-7 cells. Therefore, we report a novel lectin from the prawn haemolymph with high specificity for L-fucose and antiproliferative towards human cancer cells. However, further establishment of the modus operandi of this lectin is required to enable its biotechnological applications.
Collapse
Affiliation(s)
- Biji Chatterjee
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasaragod, Kerala, 671314, India
| | - Krishna Ghosh
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasaragod, Kerala, 671314, India
| | - Nitin Yadav
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasaragod, Kerala, 671314, India
| | - Santosh R Kanade
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasaragod, Kerala, 671314, India
| |
Collapse
|
18
|
Li G, Gao Y, Cui L, Wu L, Yang X, Chen J. Anguilla japonicalectin 1 delivery through adenovirus vector induces apoptotic cancer cell death through interaction with PRMT5. J Gene Med 2016; 18:65-74. [DOI: 10.1002/jgm.2878] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/20/2016] [Accepted: 03/07/2016] [Indexed: 12/22/2022] Open
Affiliation(s)
- Gongchu Li
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou Zhejiang China
| | - Yajun Gao
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou Zhejiang China
| | - Lianzhen Cui
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou Zhejiang China
| | - Liqin Wu
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou Zhejiang China
| | - Xinyan Yang
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou Zhejiang China
| | - Jing Chen
- Institute of Life Sciences, College of Life and Environmental Sciences; Hangzhou Normal University; Hangzhou Zhejiang China
| |
Collapse
|
19
|
MytiLec, a Mussel R-Type Lectin, Interacts with Surface Glycan Gb3 on Burkitt's Lymphoma Cells to Trigger Apoptosis through Multiple Pathways. Mar Drugs 2015; 13:7377-89. [PMID: 26694420 PMCID: PMC4699244 DOI: 10.3390/md13127071] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/30/2015] [Accepted: 12/02/2015] [Indexed: 12/17/2022] Open
Abstract
MytiLec; a novel lectin isolated from the Mediterranean mussel (Mytilus galloprovincialis); shows strong binding affinity to globotriose (Gb3: Galα1-4Galβ1-4Glc). MytiLec revealed β-trefoil folding as also found in the ricin B-subunit type (R-type) lectin family, although the amino acid sequences were quite different. Classification of R-type lectin family members therefore needs to be based on conformation as well as on primary structure. MytiLec specifically killed Burkitt's lymphoma Ramos cells, which express Gb3. Fluorescein-labeling assay revealed that MytiLec was incorporated inside the cells. MytiLec treatment of Ramos cells resulted in activation of both classical MAPK/ extracellular signal-regulated kinase and extracellular signal-regulated kinase (MEK-ERK) and stress-activated (p38 kinase and JNK) Mitogen-activated protein kinases (MAPK) pathways. In the cells, MytiLec treatment triggered expression of tumor necrosis factor (TNF)-α (a ligand of death receptor-dependent apoptosis) and activation of mitochondria-controlling caspase-9 (initiator caspase) and caspase-3 (activator caspase). Experiments using the specific MEK inhibitor U0126 showed that MytiLec-induced phosphorylation of the MEK-ERK pathway up-regulated expression of the cyclin-dependent kinase inhibitor p21, leading to cell cycle arrest and TNF-α production. Activation of caspase-3 by MytiLec appeared to be regulated by multiple different pathways. Our findings, taken together, indicate that the novel R-type lectin MytiLec initiates programmed cell death of Burkitt's lymphoma cells through multiple pathways (MAPK cascade, death receptor signaling; caspase activation) based on interaction of the lectin with Gb3-containing glycosphingolipid-enriched microdomains on the cell surface.
Collapse
|
20
|
Yau T, Dan X, Ng CCW, Ng TB. Lectins with potential for anti-cancer therapy. Molecules 2015; 20:3791-810. [PMID: 25730388 PMCID: PMC6272365 DOI: 10.3390/molecules20033791] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/13/2015] [Accepted: 02/15/2015] [Indexed: 02/07/2023] Open
Abstract
This article reviews lectins of animal and plant origin that induce apoptosis and autophagy of cancer cells and hence possess the potential of being developed into anticancer drugs. Apoptosis-inducing lectins encompass galectins, C-type lectins, annexins, Haliotis discus discus lectin, Polygonatum odoratum lectin, mistletoe lectin, and concanavalin A, fucose-binding Dicentrarchus labrax lectin, and Strongylocentrotus purpuratus lectin, Polygonatum odoratum lectin, and mistletoe lectin, Polygonatum odoratum lectin, autophagy inducing lectins include annexins and Polygonatum odoratum lectin.
Collapse
Affiliation(s)
- Tammy Yau
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA.
| | - Xiuli Dan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| | - Charlene Cheuk Wing Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| |
Collapse
|