1
|
Tsuge H, Habuka N, Yoshida T. General ADP-Ribosylation Mechanism Based on the Structure of ADP-Ribosyltransferase-Substrate Complexes. Toxins (Basel) 2024; 16:313. [PMID: 39057953 PMCID: PMC11281641 DOI: 10.3390/toxins16070313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
ADP-ribosylation is a ubiquitous modification of proteins and other targets, such as nucleic acids, that regulates various cellular functions in all kingdoms of life. Furthermore, these ADP-ribosyltransferases (ARTs) modify a variety of substrates and atoms. It has been almost 60 years since ADP-ribosylation was discovered. Various ART structures have been revealed with cofactors (NAD+ or NAD+ analog). However, we still do not know the molecular mechanisms of ART. It needs to be better understood how ART specifies the target amino acids or bases. For this purpose, more information is needed about the tripartite complex structures of ART, the cofactors, and the substrates. The tripartite complex is essential to understand the mechanism of ADP-ribosyltransferase. This review updates the general ADP-ribosylation mechanism based on ART tripartite complex structures.
Collapse
Affiliation(s)
- Hideaki Tsuge
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 6038555, Japan
| | - Noriyuki Habuka
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 6038555, Japan
| | - Toru Yoshida
- Faculty of Sciences, Japan Women’s University, Tokyo 1120015, Japan
| |
Collapse
|
2
|
Zheng B, Wang G, Qu Z, Hu J, Bao Z, Wang M. Glycosaminoglycan lyase: A new competition between bacteria and the pacific white shrimp Litopenaeus vannamei. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 156:105177. [PMID: 38593892 DOI: 10.1016/j.dci.2024.105177] [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: 01/14/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Horizontal gene transfer (HGT) is an important evolutionary force in the formation of prokaryotic and eukaryotic genomes. In recent years, many HGT genes horizontally transferred from prokaryotes to eukaryotes have been reported, and most of them are present in arthropods. The Pacific white shrimp Litopenaeus vannamei, an important economic species of arthropod, has close relationships with bacteria, providing a platform for horizontal gene transfer (HGT). In this study, we analyzed bacteria-derived HGT based on a high-quality genome of L. vannamei via a homology search and phylogenetic analysis, and six HGT genes were identified. Among these six horizontally transferred genes, we found one gene (LOC113799989) that contains a bacterial chondroitinase AC structural domain and encodes an unknown glycosaminoglycan (GAG) lyase in L. vannamei. The real-time quantitative PCR results showed that the mRNA expression level of LOC113799989 was highest in the hepatopancreas and heart, and after stimulation by Vibrio parahaemolyticus, its mRNA expression level was rapidly up-regulated within 12 h. Furthermore, after injecting si-RNA and stimulation by V. parahaemolyticus, we found that the experimental group had a higher cumulative mortality rate in 48 h than the control group, indicating that the bacteria-derived GAG lyase can reduce the mortality of shrimp with respect to infection by V. parahaemolyticus and might be related to the resistance of shrimp to bacterial diseases. Our findings contribute to the study of the function of GAGs and provide new insights into GAG-related microbial pathogenesis and host defense mechanisms in arthropods.
Collapse
Affiliation(s)
- Bo Zheng
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Gengzhuo Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Zhe Qu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China.
| | - Jingjie Hu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
| | - Mengqiang Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China.
| |
Collapse
|
3
|
Takahashi-Nakaguchi A, Horiuchi Y, Yamamoto M, Totsuka Y, Wakabayashi K. Pierisin, Cytotoxic and Apoptosis-Inducing DNA ADP-Ribosylating Protein in Cabbage Butterfly. Toxins (Basel) 2024; 16:270. [PMID: 38922164 PMCID: PMC11209040 DOI: 10.3390/toxins16060270] [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: 05/11/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Pierisin-1 was serendipitously discovered as a strong cytotoxic and apoptosis-inducing protein from pupae of the cabbage butterfly Pieris rapae against cancer cell lines. This 98-kDa protein consists of the N-terminal region (27 kDa) and C-terminal region (71 kDa), and analysis of their biological function revealed that pierisin-1 binds to cell surface glycosphingolipids on the C-terminal side, is taken up into the cell, and is cleaved to N- and C-terminal portions, where the N-terminal portion mono-ADP-ribosylates the guanine base of DNA in the presence of NAD to induce cellular genetic mutation and apoptosis. Unlike other ADP-ribosyltransferases, pieisin-1 was first found to exhibit DNA mono-ADP-ribosylating activity and show anti-cancer activity in vitro and in vivo against various cancer cell lines. Pierisin-1 was most abundantly produced during the transition from the final larval stage to the pupal stage of the cabbage butterfly, and this production was regulated by ecdysteroid hormones. This suggests that pierisn-1 might play a pivotal role in the process of metamorphosis. Moreover, pierisin-1 could contribute as a defense factor against parasitization and microbial infections in the cabbage butterfly. Pierisin-like proteins in butterflies were shown to be present not only among the subtribe Pierina but also among the subtribes Aporiina and Appiadina, and pierisin-2, -3, and -4 were identified in these butterflies. Furthermore, DNA ADP-ribosylating activities were found in six different edible clams. Understanding of the biological nature of pierisin-1 with DNA mono-ADP-ribosylating activity could open up exciting avenues for research and potential therapeutic applications, making it a subject of great interest in the field of molecular biology and biotechnology.
Collapse
Affiliation(s)
| | - Yu Horiuchi
- Aquatic Food Research Laboratory, Central Research Institute, Tokyo Innovation Center, Nissui Corporation, 1-32-3 Shichikoku, Hachioji City 192-0991, Japan
| | - Masafumi Yamamoto
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yukari Totsuka
- Department of Environmental Health Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Keiji Wakabayashi
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
4
|
Sahoo CR, Paidesetty SK, Sarathbabu S, Dehury B, Senthil Kumar N, Padhy RN. Molecular dynamics simulation, synthesis and topoisomerase inhibitory actions of vanillin derivatives: a systematic computational structural integument. J Biomol Struct Dyn 2022; 40:11653-11663. [PMID: 34355674 DOI: 10.1080/07391102.2021.1961867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A series of 4-hydroxy-3-methoxy benzaldehyde (vanillin) derivatives (3a-3r) was designed for the principle of Schiff base condensation with several individual sulfanilamide analogues. The inhibitory potencies of the designed compounds were evaluated through molecular docking simulation studies against the targets, breast cancer-topo isomerase-IIα and estrogen receptor-α; and the top scoring poses with higher binding energy were selected to assess the mode of binding and stability of each complex through molecular dynamics simulations. Compounds that remained stable in the active sites of the both target receptors through a number of strong H-bonds and hydrophobic contacts were selected. Based on the computational results, these selected compounds, 3b, 3e and 3f were synthesized and were followed up for structural elucidation attempts, by FT/ATR, 1H NMR and 13C NMR. From the experimental in vitro studies on 3b, 3e and 3f, the following remarkable activities against breast cancer cell line were done; IC50 values of 3b, 3e and 3f were noted, 6.7, 4.3 and 11 ng/mL, respectively. These newly synthesized compounds may be used as novel inhibitors of nuclear receptors with potential therapeutic applications in control of cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chita Ranjan Sahoo
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India.,Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sudhir Kumar Paidesetty
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | | | - Budheswar Dehury
- Biomedical Informatics Centre, ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India
| | | | - Rabindra Nath Padhy
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| |
Collapse
|
5
|
Zhang Q, Kandasamy K, Alyami NM, Alyami HM, Natarajan N, Elayappan PK. Influence of Padina gymnospora on Apoptotic Proteins of Oral Cancer Cells-a Proteome-Wide Analysis. Appl Biochem Biotechnol 2022; 194:5945-5962. [PMID: 35849254 DOI: 10.1007/s12010-022-04045-w] [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] [Accepted: 06/24/2022] [Indexed: 11/25/2022]
Abstract
Oral carcinoma is one of the most vicious forms of cancer with a very low survival rate, as its patients often respond poorly to conventional chemotherapy. Presently several researchers are attempting to pursue an alternative to this therapy using natural products. Considering the promising strategy and induction of apoptosis to target the cancer cells, we evaluated the influence of a seaweed Padina gymnospora (15 µg/ml and 20 µg/ml) in enhancing apoptosis of oral cancer cells (KB-CHR-8-5) after 24-h incubation. The morphological changes indicating apoptosis were primarily assessed using a light microscope after which the apoptosis was confirmed by performing AO/EB staining method. Subsequently, MMP and ROS levels in the cells were assessed using Rh 123 and DCFH-DA staining procedures, respectively. All the above tests confirmed the ability of P. gymnospora to accelerate apoptosis in the oral cancer cells. As a next step, wide proteome analysis was performed where the proteins from P. gymnospora-treated cells were separated using the 2D electrophoresis technique and compared with that of control cells to isolate the differentially expressed proteins. This procedure resulted in the isolation of 10 proteins which were identified using MALDI-TOF/TOF MS, which established that most of the isolated proteins were part of the apoptotic process of the cell. The proteins identified are part of huge and complex pathways where it gets linked with many more genes which are also associated with apoptosis. Bioinformatics of these identified proteins was analyzed using STRING and PANTHER databases. These proteins contribute to cell apoptosis by affecting various functions, biological processes, and the synthesis of cellular components. PANTHER also demonstrated that these proteins belong to the classes of proteins that take part in several vital pathways of the cell among which the apoptotic pathway is the predominant one.
Collapse
Affiliation(s)
- Qian Zhang
- School of Stomatology, QiLu Medical University, No.1678, Renmin West Road, Zibo City, 255300, China
| | - Kavitha Kandasamy
- Department of Biochemistry, Vivekanandha College of Arts and Sciences for Women (Autonomous), Elayampalayam, Tiruchengode, Namakkal, 637205, India
| | - Nouf M Alyami
- Department of Zoology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| | - Hanadi M Alyami
- Dental Administration, King Fahad Medical City, Riyadh, 11451, Saudi Arabia
| | - Nandakumar Natarajan
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Poorni Kaliyappan Elayappan
- Department of Biochemistry, Vivekanandha College of Arts and Sciences for Women (Autonomous), Elayampalayam, Tiruchengode, Namakkal, 637205, India.
| |
Collapse
|
6
|
Elderdery AY, Alhamidi AH, Elkhalifa AME, Althobiti MM, Eltayeb Omer N, Alsugoor MH, Alsuhaymi N, Atebien EM, Hamza SMA, Alzahrani B, Alanazi F, Subbiah SK, Mok PL. Synthesis, Characterization, and Antimicrobial and Antiproliferative Effects of CuO-TiO 2-Chitosan-Escin Nanocomposites on Human Leukemic MOLT4 Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12213753. [PMID: 36364538 PMCID: PMC9655830 DOI: 10.3390/nano12213753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/15/2022] [Accepted: 10/19/2022] [Indexed: 05/13/2023]
Abstract
Nanocomposites comprised of CuO-TiO2-chitosan-escin, which has adjustable physicochemical properties, provide a solution for therapeutic selectivity in cancer treatment. By controlling the intrinsic signaling primarily through the mitochondrial signaling pathway, we desired nanocomposites with enhanced anticancer activity by containing CuO-TiO2-chitosan-escin. The metal oxides CuO and TiO2, the natural polymer chitosan, and a phytochemical compound escin were combined to form CuO-TiO2-chitosan-escin nanocomposites. The synthesized nanocomposites were confirmed and characterized using FTIR spectroscopy, TEM, and UV-Vis absorption spectroscopy. A human leukemia cell line (MOLT-4) was used to assess the efficacy and selectivity of nanocomposites. Based on a cytotoxicity study, CuO-TiO2-chitosan-escin nanocomposites had inhibition concentrations (IC50) of 13.68, 8.9, and 7.14 µg/mL against human T lymphoblast cells after 24, 48, and 72 h of incubation, respectively. Compared with untreated MOLT-4 cells, CuO-TiO2-chitosan-escin nanocomposite-treated cells significantly increased (p < 0.05) caspase-3, -8, and -9 and decreased the levels of antioxidant enzymes GR, SOD, and GSH. Furthermore, MDA for lipid peroxidase and ROS levels significantly increased (p < 0.05) in the treated cells than in the untreated cells. Remarkably, CuO-TiO2-chitosan-escin nanocomposite-mediated control of cell cycles were mainly achieved through the activation of caspase-3, -8, and -9.
Collapse
Affiliation(s)
- Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia
- Health Sciences Research Unit, Jouf University, Sakaka 42421, Saudi Arabia
- Correspondence: (A.Y.E.); (P.L.M.)
| | - Abdulaziz H. Alhamidi
- Clinical Laboratory Sciences Department, College of Applied Medical Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed M. E. Elkhalifa
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh 13316, Saudi Arabia
| | - Maryam M. Althobiti
- Department of Clinical Laboratory Science, College of Applied Medical Science, King Saud University, Shaqra 15572, Saudi Arabia
| | | | - Mahdi H. Alsugoor
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia
| | - Naif Alsuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia
| | - Entesar M. Atebien
- Department of Clinical Laboratory Science, College of Applied Medical Science, King Saud University, Shaqra 15572, Saudi Arabia
| | - Siddiqa M. A. Hamza
- College of Medicine, Department of Pathology, Umm Alqura University Algunfuda, Mecca 24382, Saudi Arabia
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia
| | - Fehaid Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences-AlQurayyat, Jouf University, Sakaka 42421, Saudi Arabia
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai 600073, India
| | - Pooi Ling Mok
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Correspondence: (A.Y.E.); (P.L.M.)
| |
Collapse
|
7
|
Elderdery AY, Alzahrani B, Hamza SMA, Mostafa-Hedeab G, Mok PL, Subbiah SK. Synthesis of Zinc Oxide (ZnO)-Titanium Dioxide (TiO 2)-Chitosan-Farnesol Nanocomposites and Assessment of Their Anticancer Potential in Human Leukemic MOLT-4 Cell Line. Bioinorg Chem Appl 2022; 2022:5949086. [PMID: 36212987 PMCID: PMC9534707 DOI: 10.1155/2022/5949086] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 08/11/2022] [Indexed: 12/05/2022] Open
Abstract
Leukemia is the most prevalent cancer in children and one of the most common and deadly cancers that affect adults. Several metal oxide nanoparticles, biopolymers, and phytochemicals have been discovered to target cancer cells selectively while inflicting low to no damage to healthy cells. Among the existing nanoparticle synthesis methodologies, biologically synthesized nanoparticles using phytochemicals have emerged as a straightforward, economical, and environmentally sound strategy. The synergistic antitumor potential of ZnO-TiO2-chitosan-farnesol nanocomposites (NCs) against leukemia MOLT-4 cells was investigated in the current study. After synthesizing the NCs, characterization of the same was carried out using XRD, DLS, FESEM, TEM, PL, EDX, and FTIR spectroscopy. To analyze its anticancer activity, MOLT-4 cells were cultured and treated at diverse dosages of NCs. The cell viability upon treatment was examined by MTT assay. The morphological and nuclear modifications were observed by dual staining. ROS and MMP levels were observed by DCFH-DA staining and Rh-123 dye, respectively. Furthermore, the caspase 3, 8, and 9 levels were examined by performing ELISA. The XRD patterns exhibited a hexagonal structure of the NCs. In the DLS spectrum, the hydrodynamic diameter of the NCs was observed to be 126.2 nm. The electrostatic interface between the ZnO-TiO2-chitosan-farnesol NCs was confirmed by the FTIR spectra. A significant loss of cell viability in a dosage-dependent trend confirmed the cytotoxic effect of the NCs. An elevated ROS level and MMP depletion suggested apoptosis-associated cell death via the intrinsic pathway, which was confirmed by elevated expressions of caspase 3, 8, and 9 markers. Thus, the results showed that the synthesized NCs demonstrated a remarkable anticancer potential against leukemic cells and can be potentially valuable in cancer treatments. The findings from this study conclude that this is a new approach for modifying the physicochemical characteristics of ZnO-TiO2-chitosan-farnesol composites to increase their properties and synergistically exhibit anticancer properties in human leukemic cancer cells.
Collapse
Affiliation(s)
- Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Siddiqa M. A. Hamza
- Faculty of Medicine, Department of Pathology, Umm Alqura University Algunfuda, Mecca, Saudi Arabia
| | - Gomaa Mostafa-Hedeab
- Pharmacology & Therapeutic Department, Medical College, Jouf University, Sakaka, Saudi Arabia
| | - Pooi Ling Mok
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
8
|
Elderdery AY, Alzahrani B, Hamza SMA, Mostafa-Hedeab G, Mok PL, Subbiah SK. CuO-TiO 2-Chitosan-Berbamine Nanocomposites Induce Apoptosis through the Mitochondrial Pathway with the Expression of P53, BAX, and BCL-2 in the Human K562 Cancer Cell Line. Bioinorg Chem Appl 2022; 2022:9602725. [PMID: 36164585 PMCID: PMC9509271 DOI: 10.1155/2022/9602725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
In this study, cells from human Chronic Myelogenous Leukemia (K562) were cultivated with CuO-TiO2-Chitosan-Berbamine nanocomposites. We examined nanocomposites using XRD, DLS, FESEM, TEM, PL, EDAX, and FTIR spectroscopy, as well as MTT for cytotoxicity, and AO/EtBr for apoptotic morphology assessment. The rate of apoptosis and cell cycle arrests was determined using flow cytometry. Flow cytometry was also employed to identify pro- and antiapoptotic proteins such as Bcl2, Bad, Bax, P53, and Cyt C. The FTIR spectrum revealed that the CuO-TiO2-Chitosan-Berbamine nanocomposites were electrostatically interlocked. The nanocomposites' XRD signals revealed a hexagonal shape. In the DLS spectrum, nanocomposites were found to have a hydrodynamic diameter. As a result of their cytotoxic action, nanocomposites displayed concentration-dependent cytotoxicity. The nanocomposites, like Doxorubicin, caused cell cycle phase arrest in K562 cells. After treatment with IC50 concentrations of CuO-TiO2-Chitosan-Berbamine nanocomposites and Doxorubicin, a substantial percentage of cells were in G2/M stage arrest. Caspase-3, -7, -8, -9, Bax, Bad, Cyt C, and P53 expression were considerably enhanced in K562 cells, whereas Bcl2 expression was decreased, indicating that these cells may have therapeutic potential against human blood cancer/leukemia-derived disorders. As a result, the nanocomposites demonstrated outstanding anticancer potential against leukemic cells. CuO-TiO2-Chitosan-Berbamine, according to our findings.
Collapse
Affiliation(s)
- Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Siddiqa M. A Hamza
- Faculty of Medicine, Department of Pathology, Umm Alqura University, Algunfuda, Mecca, Saudi Arabia
| | - Gomaa Mostafa-Hedeab
- Pharmacology & Therapeutic Department-Medical College, Jouf University, Sakaka, Saudi Arabia
| | - Pooi Ling Mok
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
9
|
Elderdery AY, Alzahrani B, Alanazi F, Hamza SM, Elkhalifa AM, Alhamidi AH, Alabdulsalam AA, Mohamedain A, Kumar SS, Mok PL. Amelioration of human acute lymphoblastic leukemia (ALL) cells by ZnO-TiO2-Chitosan-Amygdalin nanocomposites. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
10
|
Fibroin gene expression and antioxidant enzymes are elevated in Bombyx mori when reared on preferred host plants. Biologia (Bratisl) 2020. [DOI: 10.2478/s11756-020-00499-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
Sarathbabu S, Sarma RJ, Lalhruaitluanga H, Velmurugan D, Subramanian S, Kumar NS. In vitro DNA binding activity and molecular docking reveals pierisin-5 as an anti-proliferative agent against gastric cancer. J Biomol Struct Dyn 2019; 38:3474-3488. [PMID: 31530224 DOI: 10.1080/07391102.2019.1659856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Pierisin-5 is a DNA dependent ADP ribosyltransferase (ADRT) protein from the larvae of Indian cabbage white butterfly, Pieris canidia. Interestingly, Pierisin-5 ADP-ribosylates the DNA as a substrate, but not the protein and subsequently persuades apoptotic cell death in human cancer cells. This has led to the investigation on the DNA binding activity of Pierisin-5 using in vitro and in silico approaches in the present study. However, both the structure and the mechanism of ADP-ribosylation of pierisin-5 are unknown. In silico modeled structure of the N-terminal ADRT catalytic domain interacted with the minor groove of B-DNA for ribosylation with the help of β-NAD+ which lead to a structural modification in DNA (DNA adduct). The possible interaction between calf thymus DNA (CT-DNA) and purified pierisin-5 protein was studied through spectral-spatial studies and the blue shift and hyperchromism in the UV-Visible spectra was observed. The DNA adduct property of pierisin-5 protein was validated by in vitro cytotoxic assay on human gastric (AGS) cancer cell lines. Our study is the first report of the mechanism of DNA binding property of pierisin-5 protein which leads to the induction of cytotoxicity and apoptotic cell death against cancer cell lines.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | | | - Devadasan Velmurugan
- Centre of Advanced Studies in Crystallography and Biophysics, University of Madras, Chennai, India
| | - Selvi Subramanian
- Department of Biotechnology, PSG College of Technology, Coimbatore, India
| | | |
Collapse
|
12
|
Li H, Chen LP, Wang T, Wang SG, Liu JH. Calpain inhibition improves erectile function in diabetic mice via upregulating endothelial nitric oxide synthase expression and reducing apoptosis. Asian J Androl 2019; 20:342-348. [PMID: 29319007 PMCID: PMC6038160 DOI: 10.4103/aja.aja_63_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calpain activation contributes to hyperglycemia-induced endothelial dysfunction and apoptosis. This study was designed to investigate the role of calpain inhibition in improving diabetic erectile dysfunction (ED) in mice. Thirty-eight-week-old male C57BL/6J mice were divided into three groups: (1) nondiabetic control group, (2) diabetic mice + vehicle group, and (3) diabetic mice + MDL28170 (an inhibitor of calpain) group. Type 1 diabetes was induced by intraperitoneal injection of streptozotocin at 60 mg kg−1 body weight for 5 consecutive days. Thirteen weeks later, diabetic mice were treated with MDL28170 or vehicle for 4 weeks. The erectile function was assessed by electrical stimulation of the cavernous nerve. Penile tissues were collected for measurement of calpain activity and the endothelial nitric oxide synthase (eNOS)-nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) pathway. Terminal deoxynucleotidyl transferase 2'-deoxyuridine 5'-triphosphate nick end labeling (TUNEL) staining was used to evaluate apoptosis. Caspase-3 expression and activity were also measured to determine apoptosis. Our results showed that erectile function was enhanced by MDL28170 treatment in diabetic mice compared with the vehicle diabetic group. No differences in calpain-1 and calpain-2 expressions were observed among the three groups. However, calpain activity was increased in the diabetic group and reduced by MDL28170. The eNOS-NO-cGMP pathway was upregulated by MDL28170 treatment in diabetic mice. Additionally, MDL28170 could attenuate apoptosis and increase the endothelium and smooth muscle levels in corpus cavernosum. Inhibition of calpain could improve erectile function, probably by upregulating the eNOS-NO-cGMP pathway and reducing apoptosis.
Collapse
Affiliation(s)
- Hao Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li-Ping Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shao-Gang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ji-Hong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
13
|
Subbarayan S, Subramanian S, Senthil Kumar N. Recombinant Pierisin-5 Induces Apoptosis and Differential Expression of Bcl-2, Bax, and p53 in Human Cancer Cells. DNA Cell Biol 2019; 38:773-785. [DOI: 10.1089/dna.2018.4520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
| | - Selvi Subramanian
- Department of Biotechnology, PSG College of Technology, Coimbatore, India
| | | |
Collapse
|
14
|
Sarathbabu S, Marimuthu SK, Ghatak S, Vidyalakshmi S, Gurusubramanian G, Ghosh SK, Subramanian S, Zhang W, Kumar NS. Induction of Apoptosis by Pierisin-6 in HPV Positive HeLa and HepG2 Cancer Cells is Mediated by the Caspase-3 Dependent Mitochondrial Pathway. Anticancer Agents Med Chem 2019; 19:337-346. [DOI: 10.2174/1871520619666181127113848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/18/2018] [Accepted: 11/14/2018] [Indexed: 11/22/2022]
Abstract
Background:
To explore the cytotoxic and apoptotic activity of the pierisin-6 protein in HPV HeLa
and HepG2 cell lines.
Methods:
In this study, isolation, and purification of cytotoxic Prierisin-6 from the larvae of Pieris napi by
affinity column chromatography techniques. Characterization of full-length mRNA of pierisin-6 gene was performed
using 3’/5’ RACE PCR. The quantitative RT-PCR used to study the developmental stage-specific expression
of pierisin-6 mRNA. The most effective concentration of Pierisin-6 protein was determined by measuring
cell proliferation. Apoptosis was assessed using AO/Et-Br, Propidium Iodide, and Rhodamine 123 assays,
whereas protein levels of caspase 3, cytochrome C were evaluated by ELISA method. Pierisin-6 induced cell
cycle arrest was determined using Propidium iodide by FACS.
Results:
In this study, Pierisin-6, a novel apoptotic protein was found to have cytotoxicity against HeLa, HepG2
human cancer cell lines and L-132 human lung epithelial cell line. Among the target cells, HeLa was the most
sensitive to Pierisin-6. Flow cytometry analysis confirms an increased percentage of apoptotic cells in sub G1
phase and cell cycle arrest at S phase. Alteration in the transmembrane potential of mitochondria, Cytochrome c
released from the mitochondrial membrane, and caspase substrate assay demonstrated the cleavage of Ac-
DEVD-pNA signifying the activation of Caspase-3. These findings suggested that Pierisin-6 significantly induce
apoptosis in HeLa and HepG2 cells and is attributed mainly through a mitochondrial pathway by activation of
caspases. The developmental and stage-specific expression of pierisin-6 mRNA was one thousand-fold increased
from second to third instar larvae and gradually declined before pupation.
Conclusion:
Pierisin-6 represents a promising therapeutic approach for liver cancer patients.
Collapse
Affiliation(s)
| | - Satheesh K. Marimuthu
- Department of Biotechnology, PSG College of Technology, Coimbatore-641004, TN, India
| | - Souvik Ghatak
- Department of Biotechnology, Mizoram University, Aizawl-796004, Mizoram, India
| | | | | | - Sankar K. Ghosh
- Department of Biotechnology, Assam University, Silchar-788011, Assam, India
| | - Selvi Subramanian
- Department of Biotechnology, PSG College of Technology, Coimbatore-641004, TN, India
| | - Wenqing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Nachimuthu S. Kumar
- Department of Biotechnology, Mizoram University, Aizawl-796004, Mizoram, India
| |
Collapse
|
15
|
Liu R, Cheng Q, Song X, Wang H, Wang X, Wang L, Zhu B, Song L. A vital ubiquitin-conjugating enzyme CgUbe2g1 participated in regulation of immune response of Pacific oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 91:132-142. [PMID: 30389518 DOI: 10.1016/j.dci.2018.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 06/08/2023]
Abstract
As an important post-translational protein modification, ubiquitination has been demonstrated to play a vital role in immune response of vertebrates. Ubiquitin (Ub)-conjugating enzyme E2 is the "heart" of ubiquitination, which is responsible for Ub cellular signaling and substrate modification. In the present study, an Ub-conjugating enzyme E2 (designed as CgUbe2g1) was identified from oyster Crassostrea gigas, and its regulation in the immune response against lipopolysaccharide (LPS) stimulation was investigated. CgUbe2g1 encoded a polypeptide of 168 amino acids with the predicted molecular mass of 19.20 kDa and contained conserved catalytic 'Ubc' domains. It shared a higher similarity with the known UBC2G1 type E2s and was closely clustered with the type E2s identified from invertebrates in the phylogenetic assay. The mRNA transcripts of CgUbe2g1 were mainly distributed in hemocyte, mantle, hepatopancreas and male gonad of C. gigas. CgUbe2g1 protein was found to be colocalized with Ub around the nucleus of oyster hemocyte. The recombinant CgUbe2g1 protein (rCgUbe2g1) could activate the ubiquitination in vitro by binding both activated and un-activated Ub. The expressions of inflammation-related factors TNF-α and NF-κB in CgUbe2g1 transfected cells were both significantly up-regulated after LPS stimulation, which were 12.9-fold at 3 h (p < 0.01) and 2.3-fold at 6 h (p < 0.01) of that in negative control group, respectively. The phagocytic rate of hemocyte and the ROS level in hemocyte were both significantly decreased (p < 0.01), while the apoptosis rate was significantly increased (p < 0.01) after CgUbe2g1 mRNA was interfered. These results demonstrated that Ub-conjugating enzyme CgUbe2g1 was involved in the innate immune response of oyster against invading pathogen, which might play important roles in the activation of inflammatory response and regulation of cellular immune response.
Collapse
Affiliation(s)
- Rui Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Qi Cheng
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Xiaorui Song
- Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Hao Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Xiudan Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Linsheng Song
- Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
16
|
Lv T, Xu L, Wu G, Li C, Wen Y, Zhang T, Gao Y, Chen H. Construction and biological evaluation of different self-assembled nanoarchitectures of FZU-03,010. Eur J Pharm Sci 2018; 121:382-391. [PMID: 29908299 DOI: 10.1016/j.ejps.2018.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/25/2018] [Accepted: 06/10/2018] [Indexed: 02/06/2023]
Abstract
Chemotherapy is currently one of the promising therapeutic methods for non-small-cell lung cancer (NSCLC), but the emergence of multidrug resistance (MDR) is the greatest obstacle to efficient drug delivery for successful chemotherapy. Nanotechnology-based drug delivery holds great promise to promote intracellular drug delivery to reverse MDR. In this work, we used our previously synthesized ursolic acid (UA) derivative, FZU-03,010 (F3), to prepare nanodrugs of F3 with different architectures and study the role of the structure on the physiochemical properties and the biological effects against A549 and its PTX-resistant A549/PTX lung cancer cells. Using different preparation methods, amphiphilic F3 could self-assemble into different structures such as nanoaggregates (F3-NA), vesicles (F3-VC), or nanoparticles (F3-NP) with different physiochemical properties. The self-assembled nanodrugs could be utilized for the entrapment of fluorophores and showed different cellular uptake efficiencies. The cytotoxicity results demonstrated that compared with UA, F3-NA and F3-NP could suppress A549 and A549/PTX cells viability more potently at lower concentration. In addition, F3-NA and F3-NP could induce G1 cell cycle arrest, cell apoptosis and caspase-3 activation more efficiently than that of UA. Furthermore, F3-NA and F3-NP could effectively inhibit PI3K/Akt pathway and decrease the expression of Bcl-2 and the cell cycle-dependent kinase inhibitors p-ERK1/2 and Cyclin D1 in both A549 and A549/PTX cells. In conclusion, our results suggest that the UA derivative F3 is more potent in inhibiting cancer cell proliferation, and F3-NA and F3-NP have the potential to be developed as a therapeutic agent for resistant NSCLC cells.
Collapse
Affiliation(s)
- Tingting Lv
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Liang Xu
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Guolin Wu
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Cailong Li
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Yibo Wen
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Tao Zhang
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Yu Gao
- College of Chemistry, Fuzhou University, Fuzhou 350108, China.
| | - Haijun Chen
- College of Chemistry, Fuzhou University, Fuzhou 350108, China.
| |
Collapse
|
17
|
Singh G, Passsari AK, Singh P, Leo VV, Subbarayan S, Kumar B, Singh BP, lalhlenmawia H, Kumar NS. Pharmacological potential of Bidens pilosa L. and determination of bioactive compounds using UHPLC-QqQ LIT-MS/MS and GC/MS. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:492. [PMID: 29145848 PMCID: PMC5689161 DOI: 10.1186/s12906-017-2000-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/10/2017] [Indexed: 12/27/2022]
Abstract
Background Research of natural products from traditionally used medicinal plants to fight against the human ailments is fetching attention of researchers worldwide. Bidens pilosa Linn. var. Radiata (Asteraceae) is well known for its folkloric medicinal use against various diseases from many decades. Mizoram, North East India, has high plant diversity and the use of this plant as herbal medicine is deep rooted in the local tribes. The present study was executed to understand the pharmacological potential of B. pilosa leaves extract. Methods The antimicrobial potential was determined using agar well diffusion and broth microdilution method against bacterial and yeast pathogens. Cytotoxicity was evaluated using MTT and apoptotic DNA fragmentation assays. Further, the antioxidant ability of the extract was analysed using DPPH and ABTS free radical scavenging assay. Mosquitocidal activity was evaluated against third in-star larvae of C. quinquefasciatus using dose response and time response larvicidal bioassay. Additionally, the major phenolic and volatile compounds were determined using UHPLC-QqQLIT-MS/MS and GC/MS respectively. Results We found that the extract showed highest antimicrobial activity against E. coli (MIC 80 μg/mL and IC50 110.04 μg/mL) and showed significant cytotoxicity against human epidermoid carcinoma (KB-3-1) cells with IC50 values of 99.56 μg/mL among the tested cancer cell lines. The IC50 values for scavenging DPPH and ABTS was 80.45 μg/mL and 171.6 μg/mL respectively. The extract also showed the high phenolics (72 μg GAE/mg extract) and flavonoids (123.3 μg Quercetin /mg extract). Lastly, five bioactive and six volatile compounds were detected using UHPLC-QqQLIT-MS/MS and GC-MS respectively which may be responsible for the plant’s bioactivities. An anticancerous compound, Paclitaxel was detected and quantified for the first time from B. pilosa leaves extract, which further showed the anticancerous potential of the tested extract. Conclusion On the basis of the present investigation, we propose that the leaf extract of B. pilosa might be a good candidate for the search of efficient environment friendly natural bioactive agent and pharmaceutically important compounds.
Collapse
|
18
|
Oda T, Hirabayashi H, Shikauchi G, Takamura R, Hiraga K, Minami H, Hashimoto H, Yamamoto M, Wakabayashi K, Shimizu T, Sato M. Structural basis of autoinhibition and activation of the DNA-targeting ADP-ribosyltransferase pierisin-1. J Biol Chem 2017; 292:15445-15455. [PMID: 28765284 DOI: 10.1074/jbc.m117.776641] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 07/25/2017] [Indexed: 01/10/2023] Open
Abstract
ADP-ribosyltransferases transfer the ADP-ribose moiety of βNAD+ to an acceptor molecule, usually a protein that modulates the function of the acceptor. Pierisin-1 is an ADP-ribosyltransferase from the cabbage butterfly Pieris rapae and is composed of N-terminal catalytic and C-terminal ricin B-like domains. Curiously, it ADP-ribosylates the DNA duplex, resulting in apoptosis of various cancer cells, which has raised interest in pierisin-1 as an anti-cancer agent. However, both the structure and the mechanism of DNA ADP-ribosylation are unclear. Here, we report the crystal structures of the N-terminal catalytic domain of pierisin-1, its complex with βNAD+, and the catalytic domain with the linker connecting it to the ricin B-like domains. We found that the catalytic domain possesses a defined, positively charged region on the molecular surface but that its overall structure is otherwise similar to those of protein-targeting ADP-ribosyltransferases. Electrophoretic mobility shift assays and site-directed mutagenesis indicated that pierisin-1 binds double-stranded but not single-stranded DNA and that Lys122, Lys123, and Lys124, which are found in a loop, and Arg181 and Arg187, located in a basic cleft near the loop, are required for DNA binding. Furthermore, the structure of the catalytic domain with the linker revealed an autoinhibitory mechanism in which the linker occupies and blocks both the βNAD+- and DNA-binding sites, suggesting that proteolytic cleavage to remove the linker is necessary for enzyme catalysis. Our study provides a structural basis for the DNA-acceptor specificity of pierisin-1 and reveals that a self-regulatory mechanism is required for its activity.
Collapse
Affiliation(s)
- Takashi Oda
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hirokazu Hirabayashi
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Gen Shikauchi
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Ryouma Takamura
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Kiyoshi Hiraga
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Minami
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Hashimoto
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.,the School of Pharmaceutical Sciences and
| | - Masafumi Yamamoto
- the Central Institute for Experimental Animals, Kawasaki 210-0821, Japan, and
| | - Keiji Wakabayashi
- the Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Toshiyuki Shimizu
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.,the Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Mamoru Sato
- From the Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan,
| |
Collapse
|
19
|
Shen J, Cong Q, Kinch LN, Borek D, Otwinowski Z, Grishin NV. Complete genome of Pieris rapae, a resilient alien, a cabbage pest, and a source of anti-cancer proteins. F1000Res 2016; 5:2631. [PMID: 28163896 PMCID: PMC5247789 DOI: 10.12688/f1000research.9765.1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2016] [Indexed: 11/20/2022] Open
Abstract
The Small Cabbage White ( Pieris rapae) is originally a Eurasian butterfly. Being accidentally introduced into North America, Australia, and New Zealand a century or more ago, it spread throughout the continents and rapidly established as one of the most abundant butterfly species. Although it is a serious pest of cabbage and other mustard family plants with its caterpillars reducing crops to stems, it is also a source of pierisin, a protein unique to the Whites that shows cytotoxicity to cancer cells. To better understand the unusual biology of this omnipresent agriculturally and medically important butterfly, we sequenced and annotated the complete genome from USA specimens. At 246 Mbp, it is among the smallest Lepidoptera genomes reported to date. While 1.5% positions in the genome are heterozygous, they are distributed highly non-randomly along the scaffolds, and nearly 20% of longer than 1000 base-pair segments are SNP-free (median length: 38000 bp). Computational simulations of population evolutionary history suggest that American populations started from a very small number of introduced individuals, possibly a single fertilized female, which is in agreement with historical literature. Comparison to other Lepidoptera genomes reveals several unique families of proteins that may contribute to the unusual resilience of Pieris. The nitrile-specifier proteins divert the plant defense chemicals to non-toxic products. The apoptosis-inducing pierisins could offer a defense mechanism against parasitic wasps. While only two pierisins from Pieris rapae were characterized before, the genome sequence revealed eight, offering additional candidates as anti-cancer drugs. The reference genome we obtained lays the foundation for future studies of the Cabbage White and other Pieridae species.
Collapse
Affiliation(s)
- Jinhui Shen
- Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, USA
| | - Qian Cong
- Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, USA
| | - Lisa N Kinch
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, USA
| | - Dominika Borek
- Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, USA
| | - Zbyszek Otwinowski
- Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, USA
| | - Nick V Grishin
- Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, USA
| |
Collapse
|