1
|
Guo H, Hong J, Zhu Y, Gui H, Liu H, Ren R, Li Y, Shan S, Guan Z, Liu M, Yang Z. A Mannosylated peptidyl lipid CManDA doped into cytidinyl/cationic lipids efficiently delivers siG12Ss to lung cancer in vivo. J Control Release 2025; 381:113624. [PMID: 40073943 DOI: 10.1016/j.jconrel.2025.113624] [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: 12/31/2024] [Revised: 03/02/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Gene therapy has attracted widespread attention in recent years, and one of the important delivery systems is the LNP. However, many LNPs have potential toxicity and accumulate in the liver. Here, we designed and synthesized a Gemini-type mannosylated peptidyl lipid called CManDA(M), which, in combination with the cytidinyl lipid DNCA(D) and the peptidyl lipid CLD(C) (D/50C/50M), could transfect siRNA (siG12S) into A549 cells to target and silence the KRASG12S gene. The fluorescence intensity in the tumor area of the D/50C/50M/Cy5.5-siG12S group increased by approximately 2.5 times. Furthermore, full 2'-F/2'-OMe-modified siG12Ss could also be transfected by D/50C/50M into cells, resulting in target gene silencing. The tumor weight in the D/50C/50M/M3 group (1.5 mg/kg, i.v.) was reduced by 50 % after administration in a mouse axillary tumor (A549) model, whereas the tumor bioluminescence intensity was only approximately 30 % of that in the blank group in a mouse orthotopic lung cancer model and showed no significant toxicity. Further studies revealed that the mannose groups of CManDA can be exposed on the nanoparticle surface to bind lectins, and CManDA can also shield the formation of a protein corona and alter the composition of the protein corona, which aids in the enhancement of its active targeting function. CManDA is expected to be a safe and effective helper lipid for tumor-targeted delivery of siRNA in vivo.
Collapse
Affiliation(s)
- Hua Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiamei Hong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuejie Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Genable (Beijing) Biotechnology Co., Ltd, (#)38 Yongda Road, Beijing 102609, China
| | - Hongzhe Gui
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hongyi Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Runan Ren
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yi Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Saijun Shan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Mingzhe Liu
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Genable (Beijing) Biotechnology Co., Ltd, (#)38 Yongda Road, Beijing 102609, China.
| |
Collapse
|
2
|
Yu Q, Xu D, Chen S, Yu Y, Yu H, Li Y, Sun W, Yin S. Cathepsin B Activatable Fluorescent Probe for Antitumor Efficiency Feedback: Attempt To Detect Certain Apoptotic Cells. Anal Chem 2025; 97:2932-2940. [PMID: 39882717 DOI: 10.1021/acs.analchem.4c05649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
As many treatments kill tumor cells by inducing apoptosis, fluorescent probes that can detect apoptosis are crucial for effective feedback regarding tumor therapy outcomes (in particular, activatable probes for better imaging). Cathepsins are enzymes that are released from lysosomes into the cytoplasm during lysosomal membrane permeabilization-induced apoptosis of many tumor cells, making them potential biomarkers of apoptotic cells. Despite their potential, to the best of our knowledge, no cathepsin-activatable fluorescent probes have been reported for this purpose. To fill this gap, we designed a cathepsin B (CTSB)-activatable fluorescent probe, A-DCO, which can distinguish tumor cells from normal cells in the free state and give feedback of enhanced fluorescence signal under drug stimulation when encapsulated into nanoparticles (NPs), enabling the evaluation of treatment efficacy. As a proof of concept, A-DCO NPs were employed rather than Annexin-V/PI double staining to detect apoptotic 4T1 and MCF-7 tumor cells via flow cytometry. In vivo experiments demonstrated that A-DCO NPs can rapidly detect apoptotic 4T1 tumor cells and are quickly metabolized after imaging, achieving timely, accurate, and safe feedback about tumor treatment effectiveness. The study offers a new tool for detecting the apoptosis of certain tumor cells and provides inspiration and direction for fluorescent probe design in the future.
Collapse
Affiliation(s)
- Qiqi Yu
- Key Laboratory of Organosilicon Chemistry and Materials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Dongdong Xu
- Key Laboratory of Organosilicon Chemistry and Materials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Sina Chen
- Key Laboratory of Organosilicon Chemistry and Materials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Yanlu Yu
- Key Laboratory of Organosilicon Chemistry and Materials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Huanan Yu
- School of Materials and Chemistry, China Jiliang University, Hangzhou 310018, P. R. China
| | - Yang Li
- Key Laboratory of Organosilicon Chemistry and Materials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Shouchun Yin
- Key Laboratory of Organosilicon Chemistry and Materials Technology of the Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| |
Collapse
|
3
|
Kusi D, Sun Y, Liu C. Advances in Manganese-based nanomaterials for cancer therapy via regulating Non-Ferrous ferroptosis. Int J Pharm 2025; 669:125101. [PMID: 39706379 DOI: 10.1016/j.ijpharm.2024.125101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Ferroptosis, a regulated form of cell death distinct from apoptosis, was first identified in 2012 and is characterized by iron-dependent lipid peroxidation driven by reactive oxygen species (ROS). Since its discovery, ferroptosis has been linked to various diseases, with recent studies highlighting its potential in cancer therapy, particularly for targeting cancer cells that are resistant to traditional treatments like chemotherapy and radiotherapy. While iron has historically been central to ferroptosis, emerging evidence indicates that non-ferrous ions, especially manganese (Mn), also play a crucial role in modulating this process. Mn-based nanomaterials have shown significant promise in cancer treatment by enhancing ROS production, depleting antioxidant defenses, and inducing ferroptosis. Additionally, these materials offer advantages in tumor imaging, immunotherapy, and catalyzing the Fenton-like reactions essential for ferroptosis. This review delves into the mechanisms of Mn-induced ferroptosis, focusing on recent advancements in Mn-based nanomaterials and their applications in chemodynamic therapy and immunotherapy. By leveraging non-ferrous ion-mediated ferroptosis, these approaches provide a novel avenue for cancer treatment. Furthermore, this review explores the potential role of Mn-based nanomaterials in the lipid metabolism pathways involved in ferroptosis and highlights the advantages of Mn ions over other metals in promoting ferroptosis. These insights offer new perspectives for the development of tumor therapies centered on Mn-based nanomaterials.
Collapse
Affiliation(s)
- Dipa Kusi
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Yan Sun
- Department of Cardiology, Zhejiang Rongjun Hospital, Jiaxing 314001 PR China.
| | - Chenguang Liu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| |
Collapse
|
4
|
Ooi VY, Yeh TY. Recent Advances and Mechanisms of Phage-Based Therapies in Cancer Treatment. Int J Mol Sci 2024; 25:9938. [PMID: 39337427 PMCID: PMC11432602 DOI: 10.3390/ijms25189938] [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: 08/01/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
The increasing interest in bacteriophage technology has prompted its novel applications to treat different medical conditions, most interestingly cancer. Due to their high specificity, manipulability, nontoxicity, and nanosize nature, phages are promising carriers in targeted therapy and cancer immunotherapy. This approach is particularly timely, as current challenges in cancer research include damage to healthy cells, inefficiency in targeting, obstruction by biological barriers, and drug resistance. Some cancers are being kept at the forefront of phage research, such as colorectal cancer and HCC, while others like lymphoma, cervical cancer, and myeloma have not been retouched in a decade. Common mechanisms are immunogenic antigen display on phage coats and the use of phage as transporters to carry drugs, genes, and other molecules. To date, popular phage treatments being tested are gene therapy and phage-based vaccines using M13 and λ phage, with some vaccines having advanced to human clinical trials. The results from most of these studies have been promising, but limitations in phage-based therapies such as reticuloendothelial system clearance or diffusion inefficiency must be addressed. Before phage-based therapies for cancer can be successfully used in oncology practice, more in-depth research and support from local governments are required.
Collapse
Affiliation(s)
| | - Ting-Yu Yeh
- Agricultural Biotechnology Laboratory, Auxergen Inc., Riti Rossi Colwell Center, 701 E Pratt Street, Baltimore, MD 21202, USA
| |
Collapse
|
5
|
Chu Y, Yuan Q, Jiang H, Wu L, Xie Y, Zhang X, Li L. A comprehensive review of the anticancer effects of decursin. Front Pharmacol 2024; 15:1303412. [PMID: 38444945 PMCID: PMC10912667 DOI: 10.3389/fphar.2024.1303412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Cancer is a globally complex disease with a plethora of genetic, physiological, metabolic, and environmental variations. With the increasing resistance to current anticancer drugs, efforts have been made to develop effective cancer treatments. Currently, natural products are considered promising cancer therapeutic agents due to their potent anticancer activity and low intrinsic toxicity. Decursin, a coumarin analog mainly derived from the roots of the medicinal plant Angelica sinensis, has a wide range of biological activities, including anti-inflammatory, antioxidant, neuroprotective, and especially anticancer activities. Existing studies indicate that decursin affects cell proliferation, apoptosis, autophagy, angiogenesis, and metastasis. It also indirectly affects the immune microenvironment and can act as a potential anticancer agent. Decursin can exert synergistic antitumor effects when used in combination with a number of common clinical anticancer drugs, enhancing chemotherapy sensitivity and reversing drug resistance in cancer cells, suggesting that decursin is a good drug combination. Second, decursin is also a promising lead compound, and compounds modifying its structure and formulation form also have good anticancer effects. In addition, decursin is not only a key ingredient in several natural herbs and dietary supplements but is also available through a biosynthetic pathway, with anticancer properties and a high degree of safety in cells, animals, and humans. Thus, it is evident that decursin is a promising natural compound, and its great potential for cancer prevention and treatment needs to be studied and explored in greater depth to support its move from the laboratory to the clinic.
Collapse
Affiliation(s)
- Yueming Chu
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Qiang Yuan
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Hangyu Jiang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Liang Wu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Yutao Xie
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China
| | - Xiaofen Zhang
- Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China
| | - Lin Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
6
|
Liu J, Ge Z, Jiang X, Zhang J, Sun J, Mao X. A comprehensive review of natural products with anti-hypoxic activity. Chin J Nat Med 2023; 21:499-515. [PMID: 37517818 DOI: 10.1016/s1875-5364(23)60410-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Indexed: 08/01/2023]
Abstract
Natural products exhibit substantial impacts in the field of anti-hypoxic traetment. Hypoxia can cause altitude sickness and other negative effect on the body. Headache, coma, exhaustion, vomiting and, in severe cases, death are some of the clinical signs. Currently, hypoxia is no longer just a concern in plateau regions; it is also one of the issues that can not be ignored by urban residents. This review covered polysaccharides, alkaloids, saponins, flavonoids, peptides and traditional Chinese compound prescriptions as natural products to protect against hypoxia. The active ingredients, effectiveness and mechanisms were discussed. The related anti-hypoxic mechanisms involve increasing the hemoglobin (HB) content, glycogen content and adenosine triphosphate (ATP) content, removing excessive reactive oxygen species (ROS), reducing lipid peroxidation, regulating the levels of related enzymes in cells, protecting the structural and functional integrity of the mitochondria and regulating the expression of apoptosis-related genes. These comprehensive summaries are beneficial to anti-hypoxic research and provide useful information for the development of anti-hypoxic products.
Collapse
Affiliation(s)
- Juncai Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Zhen Ge
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Xiao Jiang
- Medical College, Qingdao Binhai University, Qingdao 266555, China
| | - Jingjing Zhang
- Medical College, Qingdao Binhai University, Qingdao 266555, China
| | - Jianan Sun
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| | - Xiangzhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
7
|
Saeed RF, Awan UA, Saeed S, Mumtaz S, Akhtar N, Aslam S. Targeted Therapy and Personalized Medicine. Cancer Treat Res 2023; 185:177-205. [PMID: 37306910 DOI: 10.1007/978-3-031-27156-4_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Targeted therapy and personalized medicine are novel emerging disciplines of cancer research intended for treatment and prevention. One of the most significant advancements in modern oncology is the shift from an organ-centric strategy to a personalized strategy guided by deep molecular analysis. This shift in view, which focuses on the tumour's precise molecular changes, has paved the way for individualized treatment. Researchers and clinicians are using targeted therapies to select the best treatment available based on the molecular characterization of malignant cancer. In the treatment of a cancer, personalized medicine entails the use of genetic, immunological, and proteomic profiling to provide therapeutic alternatives as well as prognostic information about cancer. In this book, targeted therapies and personalized medicine have been covered for specific malignancies, including latest FDA-approved targeted therapies and it also sheds light on effective anti-cancer regimens and drug resistance. This will help to enhance our ability to conduct individualized health planning, make early diagnoses, and choose optimal medications for each cancer patient with predictable side effects and outcomes in a quickly evolving era. Various applications and tools' capacity have been improved for early diagnosis of cancer and the growing number of clinical trials that choose specific molecular targets reflects this predicament. Nevertheless, there are several limitations that must need to be addressed. Hence, in this chapter, we will discuss recent advancements, challenges, and opportunities in personalized medicine for various cancers, with a specific emphasis on target therapies in diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rida Fatima Saeed
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| | - Uzma Azeem Awan
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | | | - Sara Mumtaz
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Nosheen Akhtar
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Shaista Aslam
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| |
Collapse
|
8
|
Studies on 1,4-Quinone Derivatives Exhibiting Anti-Leukemic Activity along with Anti-Colorectal and Anti-Breast Cancer Effects. Molecules 2022; 28:molecules28010077. [PMID: 36615273 PMCID: PMC9822417 DOI: 10.3390/molecules28010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC), breast cancer, and chronic myeloid leukemia (CML) are life-threatening malignancies worldwide. Although potent therapeutic and screening strategies have been developed so far, these cancer types are still major public health problems. Therefore, the exploration of more potent and selective new agents is urgently required for the treatment of these cancers. Quinones represent one of the most important structures in anticancer drug discovery. We have previously identified a series of quinone-based compounds (ABQ-1-17) as anti-CML agents. In the current work, ABQ-3 was taken to the National Cancer Institute (NCI) for screening to determine its in vitro antiproliferative effects against a large panel of human tumor cell lines at five doses. ABQ-3 revealed significant growth inhibition against HCT-116 CRC and MCF-7 breast cancer cells with 2.00 µM and 2.35 µM GI50 values, respectively. The MTT test also showed that ABQ-3 possessed anticancer effects towards HCT-116 and MCF-7 cells with IC50 values of 5.22 ± 2.41 μM and 7.46 ± 2.76 μM, respectively. Further experiments indicated that ABQ-3 induced apoptosis in both cell lines, and molecular docking studies explicitly suggested that ABQ-3 exhibited DNA binding in a similar fashion to previously reported compounds. Based on in silico pharmacokinetic prediction, ABQ-3 might display drug-like features enabling this compound to become a lead molecule for future studies.
Collapse
|
9
|
Karmakar AK, Hasan MS, Sreemani A, Das Jayanta A, Hasan MM, Tithe NA, Biswas P. A review on the current progress of layered double hydroxide application in biomedical sectors. THE EUROPEAN PHYSICAL JOURNAL PLUS 2022; 137:801. [DOI: 10.1140/epjp/s13360-022-02993-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/22/2022] [Indexed: 01/06/2025]
|
10
|
iPCD: A Comprehensive Data Resource of Regulatory Proteins in Programmed Cell Death. Cells 2022; 11:cells11132018. [PMID: 35805101 PMCID: PMC9265749 DOI: 10.3390/cells11132018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023] Open
Abstract
Programmed cell death (PCD) is an essential biological process involved in many human pathologies. According to the continuous discovery of new PCD forms, a large number of proteins have been found to regulate PCD. Notably, post-translational modifications play critical roles in PCD process and the rapid advances in proteomics have facilitated the discovery of new PCD proteins. However, an integrative resource has yet to be established for maintaining these regulatory proteins. Here, we briefly summarize the mainstream PCD forms, as well as the current progress in the development of public databases to collect, curate and annotate PCD proteins. Further, we developed a comprehensive database, with integrated annotations for programmed cell death (iPCD), which contained 1,091,014 regulatory proteins involved in 30 PCD forms across 562 eukaryotic species. From the scientific literature, we manually collected 6493 experimentally identified PCD proteins, and an orthologous search was then conducted to computationally identify more potential PCD proteins. Additionally, we provided an in-depth annotation of PCD proteins in eight model organisms, by integrating the knowledge from 102 additional resources that covered 16 aspects, including post-translational modification, protein expression/proteomics, genetic variation and mutation, functional annotation, structural annotation, physicochemical property, functional domain, disease-associated information, protein–protein interaction, drug–target relation, orthologous information, biological pathway, transcriptional regulator, mRNA expression, subcellular localization and DNA and RNA element. With a data volume of 125 GB, we anticipate that iPCD can serve as a highly useful resource for further analysis of PCD in eukaryotes.
Collapse
|
11
|
Geng S, Gu L, Zhong L, Xu T, Sun Y. Genomic organization, evolution and functional characterization of caspase-2 and caspase-8 in miiuy croaker (Miichthys miiuy). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104308. [PMID: 34742824 DOI: 10.1016/j.dci.2021.104308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 06/13/2023]
Abstract
As the central link and executor of cell apoptosis, the caspase protease family has received extensive attention in recent years. However, the genetic characteristics and immune functions of some caspases are still unknown in fish. In our study, we cloned the full-length caspase-2 (mmCasp2) and caspase-8 (mmCasp2) of miiuy croaker, then we analyzed characteristics and functions of these two genes which are upstream of the apoptosis cascade reaction. Mmcasp2 and mmCasp8 exhibited a conserved domain (CASc), and the different part is that the mmCasp2 has a CARD domain, while mmCasp8 have two DED domains. Sequence and evolution analysis results showed that caspase-2 is more conservative than caspae-8 in the process of evolution. Cellular localization analysis showed that the distribution of mmCasp2 and mmCasp2 was in cytoplasm. The real-time PCR analysis showed that these two caspases are constitutively expressed in different tissues, and the expression of mmCasp2 and mmCasp8 were up-regulated in the liver, spleen, and kidney after infection with V. anguillarum. Lastly, qRT-PCR and Luciferase assays analysis showed that mmCasp2 and mmCasp8 can inhibit the NF-кB pathway. In general, we systematically analyzed the structure, evolution and related functional experiments of the caspase-2 and caspase-8 in miiuy croaker, which will help further understand the role caspase family plays in the apoptosis and immune response.
Collapse
Affiliation(s)
- Shang Geng
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Liping Gu
- Department of Medical Ultrasound, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lichang Zhong
- Department of Medical Ultrasound, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China.
| |
Collapse
|
12
|
In Vitro and In Silico Study of Analogs of Plant Product Plastoquinone to Be Effective in Colorectal Cancer Treatment. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030693. [PMID: 35163957 PMCID: PMC8839215 DOI: 10.3390/molecules27030693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Plants have paved the way for the attainment of molecules with a wide-range of biological activities. However, plant products occasionally show low biological activities and/or poor pharmacokinetic properties. In that case, development of their derivatives as drugs from the plant world has been actively performed. As plant products, plastoquinones (PQs) have been of high importance in anticancer drug design and discovery; we have previously evaluated and reported the potential cytotoxic effects of a series of PQ analogs. Among these analogs, PQ2, PQ3 and PQ10 were selected for National Cancer Institute (NCI) for in vitro screening of anticancer activity against a wide range of cancer cell lines. The apparent superior anticancer potency of PQ2 on the HCT-116 colorectal cancer cell line than that of PQ3 and PQ10 compared to other tested cell lines has encouraged us to perform further mechanistic studies to enlighten the mode of anti-colorectal cancer action of PQ2. For this purpose, its apoptotic effects on the HCT-116 cell line, DNA binding capacity and several crucial pharmacokinetic properties were investigated. Initially, MTT assay was conducted for PQ2 at different concentrations against HCT-116 cells. Results indicated that PQ2 exhibited significant cytotoxicity in HCT-116 cells with an IC50 value of 4.97 ± 1.93 μM compared to cisplatin (IC50 = 26.65 ± 7.85 μM). Moreover, apoptotic effects of PQ2 on HCT-116 cells were investigated by the annexin V/ethidium homodimer III staining method and PQ2 significantly induced apoptosis in HCT-116 cells compared to cisplatin. Based on the potent DNA cleavage capacity of PQ2, molecular docking studies were conducted in the minor groove of the double helix of DNA and PQ2 presented a key hydrogen bonding through its methoxy moiety. Overall, both in vitro and in silico studies indicated that effective, orally bioavailable drug-like PQ2 attracted attention for colorectal cancer treatment. The most important point to emerge from this study is that appropriate derivatization of a plant product leads to unique biologically active compounds.
Collapse
|
13
|
Selvarajoo N, Stanslas J, Islam MK, Sagineedu SR, Lian HK, Lim JCW. Pharmacological Modulation of Apoptosis and Autophagy in Pancreatic Cancer Treatment. Mini Rev Med Chem 2022; 22:2581-2595. [PMID: 35331093 DOI: 10.2174/1389557522666220324123605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/02/2022] [Accepted: 01/21/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pancreatic cancer is a fatal malignant neoplasm with infrequent signs and symptoms until a progressive stage. In 2020, GLOBOCAN reported that pancreatic cancer accounts for 4.7% of all cancer deaths. Despite the availability of standard chemotherapy regimens for treatment, the survival benefits are not guaranteed because tumor cells become chemoresistant even due to the development of chemoresistance in tumor cells even with a short treatment course, where apoptosis and autophagy play critical roles. OBJECTIVE This review compiled essential information on the regulatory mechanisms and roles of apoptosis and autophagy in pancreatic cancer, as well as drug-like molecules that target different pathways in pancreatic cancer eradication, with an aim to provide ideas to the scientific communities in discovering novel and specific drugs to treat pancreatic cancer, specifically PDAC. METHOD Electronic databases that were searched for research articles for this review were Scopus, Science Direct, PubMed, Springer Link, and Google Scholar. The published studies were identified and retrieved using selected keywords. DISCUSSION/CONCLUSION Many small-molecule anticancer agents have been developed to regulate autophagy and apoptosis associated with pancreatic cancer treatment, where most of them target apoptosis directly through EGFR/Ras/Raf/MAPK and PI3K/Akt/mTOR pathways. The cancer drugs that regulate autophagy in treating cancer can be categorized into three groups: i) direct autophagy inducers (e.g., rapamycin), ii) indirect autophagy inducers (e.g., resveratrol), and iii) autophagy inhibitors. Resveratrol persuades both apoptosis and autophagy with a cytoprotective effect, while autophagy inhibitors (e.g., 3-methyladenine, chloroquine) can turn off the protective autophagic effect for therapeutic benefits. Several studies showed that autophagy inhibition resulted in a synergistic effect with chemotherapy (e.g., a combination of metformin with gemcitabine/ 5FU). Such drugs possess a unique clinical value in treating pancreatic cancer as well as other autophagy-dependent carcinomas.
Collapse
Affiliation(s)
- Nityaa Selvarajoo
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Mohammad Kaisarul Islam
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Sreenivasa Rao Sagineedu
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Ho Kok Lian
- Department of Pathology, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Jonathan Chee Woei Lim
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
14
|
Van der Mude A. A proposed Information-Based modality for the treatment of cancer. Biosystems 2021; 211:104587. [PMID: 34915101 DOI: 10.1016/j.biosystems.2021.104587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 11/20/2021] [Accepted: 12/01/2021] [Indexed: 11/02/2022]
Abstract
Treatment modalities for cancer involve physical manipulations such as surgery, immunology, radiation, chemotherapy or gene editing. This is a proposal for an information-based modality. This modality does not change the internal state of the cancer cell directly - instead, the cancer cell is manipulated by giving it information to instruct the cell to perform an action. This modality is based on a theory of Structure Encoding in DNA, where information about body part structure controls the epigenetic state of cells in the process of development from pluripotent cells to fully differentiated cells. It has been noted that cancer is often due to errors in morphogenetic differentiation accompanied by associated epigenetic processes. This implies a model of cancer called the Epigenetic Differentiation Model. A major feature of the Structure Encoding Theory is that the characteristics of the differentiated cell are affected by inter-cellular information passed in the tissue microenvironment, which specifies the exact location of a cell in a body part structure. This is done by exosomes that carry fragments of long non-coding RNA and transposons, which convey structure information. In the normal process of epigenetic differentiation, the information passed may lead to apoptosis due to the constraints of a particular body part structure. The proposed treatment involves determining what structure information is being passed in a particular tumor, then adding artificial exosomes that overwhelm the current information with commands for the cells to go into apoptosis.
Collapse
|
15
|
Vaughan HJ, Green JJ. Recent Advances in Gene Therapy for Cancer Theranostics. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 20:100300. [PMID: 34738046 PMCID: PMC8562678 DOI: 10.1016/j.cobme.2021.100300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There is great interest in developing gene therapies for many disease indications, including cancer. However, successful delivery of nucleic acids to tumor cells is a major challenge, and in vivo efficacy is difficult to predict. Cancer theranostics is an approach combining anti-tumor therapy with imaging or diagnostic capabilities, with the goal of monitoring successful delivery and efficacy of a therapeutic agent in a tumor. Successful theranostics must maintain a high degree of anticancer targeting and efficacy while incorporating high-contrast imaging agents that are nontoxic and compatible with clinical imaging modalities. This review highlights recent advancements in theranostic strategies, including imaging technologies and genetic engineering approaches. Graphical Abstract.
Collapse
Affiliation(s)
- Hannah J. Vaughan
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA
| | - Jordan J. Green
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA
- Departments of Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, and the Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA
| |
Collapse
|
16
|
Li C, Cui L, Zhang L, Yang L, Zhuo Y, Cui J, Cui N, Zhang S. Saikosaponin D Attenuates Pancreatic Injury Through Suppressing the Apoptosis of Acinar Cell via Modulation of the MAPK Signaling Pathway. Front Pharmacol 2021; 12:735079. [PMID: 34744719 PMCID: PMC8566544 DOI: 10.3389/fphar.2021.735079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/15/2021] [Indexed: 01/14/2023] Open
Abstract
Chronic pancreatitis (CP) is a progressive fibro-inflammatory syndrome. The damage of acinar cells is the main cause of inflammation and the activation of pancreatic stellate cells (PSCs), which can thereby possibly further aggravate the apoptosis of more acinar cells. Saikosaponind (SSd), a major active ingredient derived from Chinese medicinal herb bupleurum falcatum, which exerted multiple pharmacological effects. However, it is not clear whether SSd protects pancreatic injury of CP via regulating the apoptosis of pancreatic acinar cells. This study systematically investigated the effect of SSd on pancreatic injury of CP in vivo and in vitro. The results revealed that SSd attenuate pancreatic damage, decrease the apoptosis and suppress the phosphorylation level of MAPK family proteins (JNK1/2, ERK1/2, and p38 MAPK) significantly in the pancreas of CP rats. In addition, SSd markedly reduced the apoptosis and inflammation of pancreatic acinar AR42J cells induced by cerulein, a drug induced CP, or Conditioned Medium from PSCs (PSCs-CM) or the combination of PSCs-CM and cerulein. Moreover, SSd significantly inhibited the activated phosphorylation of JNK1/2, ERK1/2, and p38 MAPK induced by cerulein or the combination of PSCs-CM and cerulein in AR42J cells. Furthermore, SSd treatment markedly decreased the protein levels of p-JNK and p-p38 MAPK caused by PSCs-CM alone. In conclusion, SSd ameliorated pancreatic injury, suppressed AR42J inflammation and apoptosis induced by cerulein, interrupted the effect of PSCs-CM on AR42J cells inflammation and apoptosis, possibly through MAPK pathway.
Collapse
Affiliation(s)
- Caixia Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| | - Lihua Cui
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| | - Jialin Cui
- The Clinical Medicine, Tianjin Medical University, Tianjin, China
| | - Naiqiang Cui
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| | - Shukun Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, China
| |
Collapse
|
17
|
Jiang H, Li Y, Xiang X, Tang Z, Liu K, Su Q, Zhang X, Li L. Chaetocin: A review of its anticancer potentials and mechanisms. Eur J Pharmacol 2021; 910:174459. [PMID: 34464601 DOI: 10.1016/j.ejphar.2021.174459] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/17/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Chaetocin is a natural metabolite product with various biological activities and pharmacological functions isolated from Chaetomium species fungi belonging to the thiodiketopyrazines. Numerous studies have demonstrated a wide range of antitumor activities of chaetocin in vitro and in vivo. Several studies have demonstrated that chaetocin suppresses the growth and proliferation of various tumour cells by regulating multiple signalling pathways related to tumour initiation and progression, inducing cancer cell apoptosis (intrinsic and extrinsic), enhancing autophagy, inducing cell cycle arrest, and inhibiting tumour angiogenesis, invasion, and migration. The antitumor effects and molecular mechanisms of chaetocin are reviewed and analysed in this paper, and the prospective applications of chaetocin in cancer prevention and therapy are also discussed. This review aimed to summarize the recent advances in the antitumor activity of chaetocin and to provide a rationale for further exploring the potential application of chaetocin in overcoming cancer in the future.
Collapse
Affiliation(s)
- Hangyu Jiang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Yuqi Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Xiaocong Xiang
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Zhili Tang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Qiang Su
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Xiaofen Zhang
- Department of Urology, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China.
| | - Lin Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; College of Bioengineering, Chongqing University, Chongqing, China.
| |
Collapse
|
18
|
Bellotti E, Cascone MG, Barbani N, Rossin D, Rastaldo R, Giachino C, Cristallini C. Targeting Cancer Cells Overexpressing Folate Receptors with New Terpolymer-Based Nanocapsules: Toward a Novel Targeted DNA Delivery System for Cancer Therapy. Biomedicines 2021; 9:biomedicines9091275. [PMID: 34572461 PMCID: PMC8471118 DOI: 10.3390/biomedicines9091275] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023] Open
Abstract
Chemotherapeutics represent the standard treatment for a wide range of cancers. However, these agents also affect healthy cells, thus leading to severe off-target effects. Given the non-selectivity of the commonly used drugs, any increase in the selective tumor tissue uptake would represent a significant improvement in cancer therapy. Recently, the use of gene therapy to completely remove the lesion and avoid the toxicity of chemotherapeutics has become a tendency in oncotherapy. Ideally, the genetic material must be safely transferred from the site of administration to the target cells, without involving healthy tissues. This can be achieved by encapsulating genes into non-viral carriers and modifying their surface with ligands with high selectivity and affinity for a relevant receptor on the target cells. Hence, in this work we evaluate the use of terpolymer-based nanocapsules for the targeted delivery of DNA toward cancer cells. The surface of the nanocapsules is decorated with folic acid to actively target the folate receptors overexpressed on a variety of cancer cells. The nanocapsules demonstrate a good ability of encapsulating and releasing DNA. Moreover, the presence of the targeting moieties on the surface of the nanocapsules favors cell uptake, opening up the possibility of more effective therapies.
Collapse
Affiliation(s)
- Elena Bellotti
- Department of Civil and Industrial Engineering, University of Pisa, 56122 Pisa, Italy; (M.G.C.); (N.B.)
- Correspondence: (E.B.); (C.C.); Tel.: +39-(010)-28961 (E.B.); +39-(050)-2217802 (C.C.)
| | - Maria Grazia Cascone
- Department of Civil and Industrial Engineering, University of Pisa, 56122 Pisa, Italy; (M.G.C.); (N.B.)
| | - Niccoletta Barbani
- Department of Civil and Industrial Engineering, University of Pisa, 56122 Pisa, Italy; (M.G.C.); (N.B.)
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, 10143 Turin, Italy; (D.R.); (R.R.); (C.G.)
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, 10143 Turin, Italy; (D.R.); (R.R.); (C.G.)
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10143 Turin, Italy; (D.R.); (R.R.); (C.G.)
| | - Caterina Cristallini
- Department of Civil and Industrial Engineering, University of Pisa, 56122 Pisa, Italy; (M.G.C.); (N.B.)
- Institute for Chemical and Physical Processes, IPCF ss Pisa, 56122 Pisa, Italy
- Correspondence: (E.B.); (C.C.); Tel.: +39-(010)-28961 (E.B.); +39-(050)-2217802 (C.C.)
| |
Collapse
|
19
|
Chen M, Wu W, Liu D, Lv Y, Deng H, Gao S, Gu Y, Huang M, Guo X, Liu B, Zhao B, Pang Q. Evolution and Structure of API5 and Its Roles in Anti-Apoptosis. Protein Pept Lett 2021; 28:612-622. [PMID: 33319655 DOI: 10.2174/0929866527999201211195551] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/30/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Apoptosis, also named programmed cell death, is a highly conserved physiological mechanism. Apoptosis plays crucial roles in many life processes, such as tissue development, organ formation, homeostasis maintenance, resistance against external aggression, and immune responses. Apoptosis is regulated by many genes, among which Apoptosis Inhibitor-5 (API5) is an effective inhibitor, though the structure of API5 is completely different from the other known Inhibitors of Apoptosis Proteins (IAPs). Due to its high expression in many types of tumors, API5 has received extensive attention, and may be an effective target for cancer treatment. In order to comprehensively and systematically understand the biological roles of API5, we summarized the evolution and structure of API5 and its roles in anti-apoptosis in this review.
Collapse
Affiliation(s)
- Meishan Chen
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Weiwei Wu
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Dongwu Liu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yanhua Lv
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Hongkuan Deng
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Sijia Gao
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yaqi Gu
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Mujie Huang
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Xiao Guo
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Baohua Liu
- Anti-Aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Qiuxiang Pang
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| |
Collapse
|
20
|
Apoptosis-Inducing TNF Superfamily Ligands for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13071543. [PMID: 33801589 PMCID: PMC8036978 DOI: 10.3390/cancers13071543] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease with apoptosis evasion as one of its hallmarks; therefore, apoptosis induction in transformed cells seems a promising approach as a cancer treatment. TNF apoptosis-inducing ligands, which are naturally present in the body and possess tumoricidal activity, are attractive candidates. The most studied proteins are TNF-α, FasL, and TNF-related apoptosis-inducing ligand (TRAIL). Over the years, different recombinant TNF family-derived apoptosis-inducing ligands and agonists have been designed. Their stability, specificity, and half-life have been improved because most of the TNF ligands have the disadvantages of having a short half-life and affinity to more than one receptor. Here, we review the outlook on apoptosis-inducing ligands as cancer treatments in diverse preclinical and clinical stages and summarize strategies of overcoming their natural limitations to improve their effectiveness.
Collapse
|
21
|
Epigallocatechin Gallate Protects against MNNG-Induced Precancerous Lesions of Gastric Carcinoma in Rats via PI3K/Akt/mTOR Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8846813. [PMID: 33628319 PMCID: PMC7880711 DOI: 10.1155/2021/8846813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/06/2020] [Accepted: 01/16/2021] [Indexed: 12/13/2022]
Abstract
Objective To evaluate the therapeutic effect of epigallocatechin gallate (EGCG) on precancerous lesions of gastric carcinoma (PLGC) and to determine whether EGCG protects against PLGC by regulating PI3K/Akt/mTOR pathway. Methods Twenty-four male Wistar rats were randomly divided into 3 groups: normal control group (NC), PLGC model group (MC), and group of PLGC rats treated with EGCG (MC + EGCG). 1-Methyl-3-nitro-1-nitrosoguanidine (MNNG) and sodium salicylate were combined and used to establish the PLGC rat animal model. The therapeutic effect of EGCG on PLGC was evaluated by body weight and pathological lesions of gastric mucosa in PLGC rats. Quantitative polymerase chain reaction (qPCR) was applied to measure the mRNA expressions of PI3K, Akt, and mTOR. The protein expressions of cleaved caspase-3, PTEN, PI3K, p-PI3K, Akt, p-Akt, p-mTOR, and mTOR were determined by automated western immunoblotting. Results The body weight decreased in PLGC rats while EGCG significantly increased body weight. The gastric mucosa of PLGC rats exhibited the pathological lesions of atrophy, intestinal metaplasia, and atypical hyperplasia while EGCG could ameliorate the pathological lesions. EGCG could upregulate the expressions of cleaved caspase-3 and PTEN and reduce the expressions of PI3K, Akt, and mTOR. Conclusions EGCG ameliorated pathological lesions of PLGC and exerted the effect of apoptosis promotion in PLGC rats. The apoptotic pathway triggered by EGCG may be related to inhibition of PI3K/Akt/mTOR pathway. It provided a theoretical basis for the PLGC treatment and gastric cancer prevention.
Collapse
|
22
|
Song X, Liu C, Wang N, Huang H, He S, Gong C, Wei Y. Delivery of CRISPR/Cas systems for cancer gene therapy and immunotherapy. Adv Drug Deliv Rev 2021; 168:158-180. [PMID: 32360576 DOI: 10.1016/j.addr.2020.04.010] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/19/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
The clustered, regularly interspaced, short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) systems are efficient and versatile gene editing tools, which offer enormous potential to treat cancer by editing genome, transcriptome or epigenome of tumor cells and/or immune cells. A large body of works have been done with CRISPR/Cas systems for genetic modification, and 16 clinical trials were conducted to treat cancer by ex vivo or in vivo gene editing approaches. Now, promising preclinical works have begun using CRISPR/Cas systems in vivo. However, efficient and safe delivery of CRISPR/Cas systems in vivo is still a critical challenge for their clinical applications. This article summarizes delivery of CRISPR/Cas systems by physical methods, viral vectors and non-viral vectors for cancer gene therapy and immunotherapy. The prospects for the development of physical methods, viral vectors and non-viral vectors for delivery of CRISPR/Cas systems are reviewed, and promising advances in cancer treatment using CRISPR/Cas systems are discussed.
Collapse
Affiliation(s)
- Xiangrong Song
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Chao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hai Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Siyan He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
23
|
Yu L, Lai Q, Gou L, Feng J, Yang J. Opportunities and obstacles of targeted therapy and immunotherapy in small cell lung cancer. J Drug Target 2021; 29:1-11. [PMID: 32700566 DOI: 10.1080/1061186x.2020.1797050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/26/2020] [Accepted: 07/06/2020] [Indexed: 02/05/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive malignant tumour which accounts for approximately 13-15% of all newly diagnosed lung cancer cases. To date, platinum-based chemotherapy are still the first-line treatments for SCLC. However, chemotherapy resistance and systemic toxicity limit the long-term clinical outcome of first-line treatment in SCLC. Recent years, targeted therapy and immunotherapy have made great breakthrough in cancer therapy, and researchers aim to exploit both as a single agent or in combination with chemotherapy to improve the survival of SCLC patients, but limited effectiveness and the adverse events remain the major obstacles in the treatment of SCLC. To overcome these challenges for SCLC therapies, prevention and early diagnosis for this refractory disease is very important. At the same time, we should reveal more information about the pathogenesis of SCLC and the mechanism of drug resistance. Finally, new treatment strategies should also be taken into considerations, such as repurposing drug, optimising of targets, combination therapy strategies or prognostic biomarkers to enhance therapeutic effects and decrease the adverse events rates in SCLC patients. This article will review the molecular biology characteristics of SCLC and discuss the opportunities and obstacles of the current therapy for SCLC patients.
Collapse
Affiliation(s)
- Lin Yu
- The Clinical Laboratory of Mianyang Central Hospital, Mianyang, China
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qinhuai Lai
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lantu Gou
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jiafu Feng
- The Clinical Laboratory of Mianyang Central Hospital, Mianyang, China
| | - Jinliang Yang
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Hejmady S, Pradhan R, Alexander A, Agrawal M, Singhvi G, Gorain B, Tiwari S, Kesharwani P, Dubey SK. Recent advances in targeted nanomedicine as promising antitumor therapeutics. Drug Discov Today 2020; 25:2227-2244. [DOI: 10.1016/j.drudis.2020.09.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/29/2020] [Accepted: 09/26/2020] [Indexed: 12/18/2022]
|
25
|
Wang M, Dong Y, Wu J, Li H, Zhang Y, Fan S, Li D. Baicalein ameliorates ionizing radiation-induced injuries by rebalancing gut microbiota and inhibiting apoptosis. Life Sci 2020; 261:118463. [PMID: 32950576 DOI: 10.1016/j.lfs.2020.118463] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/05/2020] [Accepted: 09/13/2020] [Indexed: 12/20/2022]
Abstract
AIMS Ionizing radiation (IR) induces injuries to the hematopoietic and intestinal systems, which are the leading cause of death. Baicalein, a plant-derived flavonoid, shows anti-oxidative stress, anti-apoptosis, anti-inflammation effects in many diseases. In this study, we evaluated the effects and mechanism of baicalein on IR induced intestinal and hematopoietic injuries. MAIN METHODS Mice were divided into three groups: Control, IR and IR + Baicalein. All of mice were intraperitoneally administered with 100 mg/kg baicalein or normal saline for 1 h before IR, and then a day post-IR. The changes in intestinal structure, function and molecular expression were observed by pathological experiments and western blot. 16S rRNA gene sequencing was performed to analyze gut microbiota and further predicted metabolic pathways through Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Hematopoietic function was evaluated by peripheral blood cells count and by flow cytometry analysis of hematopoietic cells composition. KEY FINDINGS Baicalein improved intestinal structure and the ability of proliferation and regeneration after mice exposed to IR, in which the rebalance of gut microbial composition played an important role. KEGG results showed that p53-related apoptotic pathways played important roles in the composition changes of gut microbiota. Then we observed that baicalein inhibited the activation of p53 and p53 mediated mitochondrial apoptosis and death receptor apoptosis in the intestine. In addition, IR induced injuries to hematopoietic system also could be ameliorated by baicalein. SIGNIFICANCE These results provide new insights into the mechanism of baicalein and support the potential of baicalein as a radioprotective medicine.
Collapse
Affiliation(s)
- Meifang Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jing Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Hongyan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yuanyang Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
26
|
Khan A, Aljarbou AN, Aldebasi YH, Allemailem KS, Alsahli MA, Khan S, Alruwetei AM, Khan MA. Fatty Acid Synthase (FASN) siRNA-Encapsulated-Her-2 Targeted Fab'-Immunoliposomes for Gene Silencing in Breast Cancer Cells. Int J Nanomedicine 2020; 15:5575-5589. [PMID: 32801705 PMCID: PMC7415462 DOI: 10.2147/ijn.s256022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose The overexpression of Her-2 in 25–30% breast cancer cases and the crosstalk between Her-2 and fatty acid synthase (FASN) establishes Her-2 as a promising target for site-directed delivery. The present study aimed to develop the novel lipid base formulations to target and inhibit the cellular proliferation of Her-2-expressing breast cancer cells through the silencing of FASN. In order to achieve this goal, we prepared DSPC/Chol and DOPE/CHEMS immunoliposomes, conjugated with the anti-Her-2 fab’ and encapsulated FASN siRNA against breast cancer cells. Methods We evaluated the size, stability, cellular uptake and internalization of various formulations of liposomes. The antiproliferative gene silencing potential was investigated by the cell cytotoxicity, crystal violet, wound healing and Western blot analyses in Her-2+ and Her-2¯ breast cancer cells. Results The data revealed that both nanosized FASN-siRNA-encapsulated liposomes showed significantly higher cellular uptake and internalization with enhanced stability. The cell viability of Her-2+ SK-BR3 cells treated with the targeted formulation of DSPC/Chol- and DOPE/CHEMS-encapsulating FASN-siRNA reduced to 30% and 20%, respectively, whereas it was found to be 45% and 36% in MCF-7 cells. The wounds were not only failed to close but they became broader in Her-2+ cells treated with targeted liposomes of siRNA. Consequently, the amount of FASN decreased by 80% in SK-BR3 cells treated with non-targeted liposomes and it was 30% and 60% in the MCF-7 cells treated with DSPC/Chol and DOPE/CHEMS liposomes, respectively. Conclusion In this study, we developed the formulation that targeted Her-2 for the suppression of FASN and, therefore, inhibited the proliferation of breast cancer cells.
Collapse
Affiliation(s)
- Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Ahmed N Aljarbou
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Yousef H Aldebasi
- Department of Optometry, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Shamshir Khan
- Dentistry and Pharmacy College, Buraydah Private Colleges, Al-Qassim, Buraydah, Saudi Arabia
| | - Abdulmohsen M Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Masood A Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| |
Collapse
|
27
|
Standardized Saponin Extract from Baiye No.1 Tea ( Camellia sinensis) Flowers Induced S Phase Cell Cycle Arrest and Apoptosis via AKT-MDM2-p53 Signaling Pathway in Ovarian Cancer Cells. Molecules 2020; 25:molecules25153515. [PMID: 32752095 PMCID: PMC7435957 DOI: 10.3390/molecules25153515] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is considered to be one of the most serious malignant tumors in women. Natural compounds have been considered as important sources in the search for new anti-cancer agents. Saponins are characteristic components of tea (Camellia sinensis) flower and have various biological activities, including anti-tumor effects. In this study, a high purity standardized saponin extract, namely Baiye No.1 tea flower saponin (BTFS), which contained Floratheasaponin A and Floratheasaponin D, were isolated from tea (Camellia sinensis cv. Baiye 1) flowers by macroporous resin and preparative liquid chromatography. Then, the component and purity were detected by UPLC-Q-TOF/MS/MS. This high purity BTFS inhibited the proliferation of A2780/CP70 cancer cells dose-dependently, which is evidenced by the inhibition of cell viability, reduction of colony formation ability, and suppression of PCNA protein expression. Further research found BTFS induced S phase cell cycle arrest by up-regulating p21 proteins expression and down-regulating Cyclin A2, CDK2, and Cdc25A protein expression. Furthermore, BTFS caused DNA damage and activated the ATM-Chk2 signaling pathway to block cell cycle progression. Moreover, BTFS trigged both extrinsic and intrinsic apoptosis—BTFS up-regulated the expression of death receptor pathway-related proteins DR5, Fas, and FADD and increased the ratio of pro-apoptotic/anti-apoptotic proteins of the Bcl-2 family. BTFS-induced apoptosis seems to be related to the AKT-MDM2-p53 signaling pathway. In summary, our results demonstrate that BTFS has the potential to be used as a nutraceutical for the prevention and treatment of ovarian cancer.
Collapse
|
28
|
Chen T, Zhang J, Zeng H, Zhang Y, Zhang Y, Zhou X, Zhou H. Antiproliferative effects of L-asparaginase in acute myeloid leukemia. Exp Ther Med 2020; 20:2070-2078. [PMID: 32782519 PMCID: PMC7401243 DOI: 10.3892/etm.2020.8904] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/06/2019] [Indexed: 01/13/2023] Open
Abstract
The antitumor enzyme L-asparaginase (L-Asp) has commonly been used for the treatment of acute lymphoblastic leukemia. However, the effects of L-Asp on acute myeloid leukemia (AML) and their underlying mechanisms have not been fully elucidated. In the present study, the effects of L-Asp on cell proliferation and apoptosis were investigated using the AML cell lines U937, HL-60 and KG-1a. The effects of combining L-Asp with mitoxantrone (MIT) and cytarabine (Ara-c) were also analyzed. The combination of MIT and Ara-C is known as MA therapy, and is a widely used therapeutic regimen for the treatment of elderly patients with refractory AML. When applied alone, L-Asp inhibited cell proliferation and induced apoptosis in each of the cell lines tested. Furthermore, the combined use of L-Asp with MA therapy further potentiated the inhibition of cell proliferation while increasing the induction of apoptosis. These findings provide evidence for the potential antitumor effect of L-Asp in AML, and indicate that improved efficacy maybe achieved by combining L-Asp with MIT and Ara-c. This combination may provide a promising new therapeutic strategy for the treatment of AML.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Juan Zhang
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Hui Zeng
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Yue Zhang
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Yong Zhang
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Xiaohuan Zhou
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Hebing Zhou
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| |
Collapse
|
29
|
Idriss M, Hodroj MH, Fakhoury R, Rizk S. Beta-Tocotrienol Exhibits More Cytotoxic Effects than Gamma-Tocotrienol on Breast Cancer Cells by Promoting Apoptosis via a P53-Independent PI3-Kinase Dependent Pathway. Biomolecules 2020; 10:biom10040577. [PMID: 32283796 PMCID: PMC7226046 DOI: 10.3390/biom10040577] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
Studies on tocotrienols have progressively revealed the benefits of these vitamin E isoforms on human health. Beta-tocotrienol (beta-T3) is known to be less available in nature compared to other vitamin E members, which may explain the restricted number of studies on beta-T3. In the present study, we aim to investigate the anti-proliferative effects and the pro-apoptotic mechanisms of beta-T3 on two human breast adenocarcinoma cell lines MDA-MB-231 and MCF7. To assess cell viability, both cell lines were incubated for 24 and 48 h, with different concentrations of beta-T3 and gamma-T3, the latter being a widely studied vitamin E isoform with potent anti-cancerous properties. Cell cycle progression and apoptosis induction upon treatment with various concentrations of the beta-T3 isoform were assessed. The effect of beta-T3 on the expression level of several apoptosis-related proteins p53, cytochrome C, cleaved-PARP-1, Bax, Bcl-2, and caspase-3, in addition to key cell survival proteins p-PI3K and p-GSK-3 α/β was determined using western blot analysis. Beta-tocotrienol exhibited a significantly more potent anti-proliferative effect than gamma-tocotrienol on both cell lines regardless of their hormonal receptor status. Beta-T3 induced a mild G1 arrest on both cell lines, and triggered a mitochondrial stress-mediated apoptotic response in MDA-MB-231 cells. Mechanistically, beta-T3′s anti-neoplastic activity involved the downregulation of phosphorylated PI3K and GSK-3 cell survival proteins. These findings suggest that vitamin E beta-T3 should be considered as a promising anti-cancer agent, more effective than gamma-T3 for treating human breast cancer and deserves to be further studied to investigate its effects in vitro and on other cancer types.
Collapse
Affiliation(s)
- Maya Idriss
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos 36, Lebanon; (M.I.); (M.H.H.)
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut 11-5020, Lebanon;
| | - Mohammad Hassan Hodroj
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos 36, Lebanon; (M.I.); (M.H.H.)
| | - Rajaa Fakhoury
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut 11-5020, Lebanon;
| | - Sandra Rizk
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos 36, Lebanon; (M.I.); (M.H.H.)
- Correspondence: ; Tel.: +961-1786456
| |
Collapse
|
30
|
Liao S, Yang Y, Chen S, Bi Y, Huang Q, Wei Z, Qin A, Liu B. IL-24 inhibits endometrial cancer cell proliferation by promoting apoptosis through the mitochondrial intrinsic signaling pathway. Biomed Pharmacother 2020; 124:109831. [PMID: 31972354 DOI: 10.1016/j.biopha.2020.109831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Endometrial cancer is a type of malignant tumor of the female reproductive system. Preserving fertility in endometrial cancer patients is currently a formidable challenge. Interleukin-24 (IL-24) is a unique cytokine tumor suppressor gene belonging to the IL-10 cytokine family. IL-24 has broad-spectrum antitumor activity through different signaling pathways but does not affect normal cells. IL-24 gene therapy may provide a new method for the treatment of endometrial cancer. METHODS Transfection was used for gene transfer. The expression of IL-24 and related pathway proteins in endometrial cancer tissue and the Ishikawa cell line was detected by immunohistochemistry and Western blotting, respectively. The antitumor function of IL-24 was examined in vitro and in vivo. Cell proliferation was determined by CCK-8 assay, cell migration was shown by wound-healing assay, and cell invasion was detected by Transwell assay. Apoptosis was analyzed by TUNEL assay, and HE staining was performed to observe the morphology of the samples. RESULTS Immunohistochemical analysis showed different expression levels of IL-24 in human endometrial cancer tissues and normal endometrial tissues. IL-24 increased protein expression of BAX and Cytochrome C, while BCL-2, MMP-3, VEGF, Caspase-9 and Caspase-3 expression was decreased. Overexpression of IL-24 inhibited cell proliferation, migration and invasion, but increased cell apoptosis in endometrial cancer. Mechanistically, we demonstrated that IL-24 inhibited endometrial cancer cell growth by inducing cell apoptosis through the mitochondrial intrinsic signaling pathway. In addition, IL-24 inhibited tumor development by inducing cell apoptosis and inhibiting angiogenesis, as shown in xenograft tumor experiments. CONCLUSIONS Our study demonstrates the antitumor effect of IL-24 on endometrial cancer and shows that IL-24 may be a promising therapeutic gene for endometrial cancer gene therapy.
Collapse
Affiliation(s)
- Shengbin Liao
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Yihua Yang
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Saiqiong Chen
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Yin Bi
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Qiuyan Huang
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Zhiyao Wei
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Aiping Qin
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China.
| | - Bo Liu
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China.
| |
Collapse
|
31
|
Xu S, Hao L, An X, Yang G, Wang F. Emerging research topics detection with multiple machine learning models. J Informetr 2019. [DOI: 10.1016/j.joi.2019.100983] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
32
|
Senapati S, Sarkar T, Das P, Maiti P. Layered Double Hydroxide Nanoparticles for Efficient Gene Delivery for Cancer Treatment. Bioconjug Chem 2019; 30:2544-2554. [PMID: 31498987 DOI: 10.1021/acs.bioconjchem.9b00434] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The use of cationic polymer based gene delivery vectors has several limitations such as low transfection efficiency, high toxicity, and inactivation by serum. The present work provides an inorganic based nanocarrier for efficient gene delivery and a method for preparing the same through a facile coprecipitation technique. The vehicle showed high loading capacity of DNA and can release the loaded DNA in a controlled pH-responsive manner. The developed gene delivery vehicle offers remarkable protection against DNase I and also provides protection against thermal damage. This vehicle also demonstrated efficient cellular uptake performance. Transfection and expression of plasmid gene encoding GFP proteins is achieved successfully by this LDH based vehicle. More interestingly, the developed Li-Al LDH efficiently induces GFP-p53 mediated apoptosis in HeLa cells exclusively sparing the normal tissue cells like NIH-3T3. The study demonstrates the potential of the developed inorganic based nanocarrier as a promising nonviral gene vector for tumor treatment.
Collapse
Affiliation(s)
- Sudipta Senapati
- School of Materials Science and Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi 221 005 , Uttar Pradesh , India
| | - Tanmoy Sarkar
- Centre for Genetic Disorders, Institute of Science , Banaras Hindu University , Varanasi 221005 , Uttar Pradesh , India
| | - Parimal Das
- Centre for Genetic Disorders, Institute of Science , Banaras Hindu University , Varanasi 221005 , Uttar Pradesh , India
| | - Pralay Maiti
- School of Materials Science and Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi 221 005 , Uttar Pradesh , India
| |
Collapse
|
33
|
Pucci C, Martinelli C, Ciofani G. Innovative approaches for cancer treatment: current perspectives and new challenges. Ecancermedicalscience 2019; 13:961. [PMID: 31537986 PMCID: PMC6753017 DOI: 10.3332/ecancer.2019.961] [Citation(s) in RCA: 395] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Every year, cancer is responsible for millions of deaths worldwide and, even though much progress has been achieved in medicine, there are still many issues that must be addressed in order to improve cancer therapy. For this reason, oncological research is putting a lot of effort towards finding new and efficient therapies which can alleviate critical side effects caused by conventional treatments. Different technologies are currently under evaluation in clinical trials or have been already introduced into clinical practice. While nanomedicine is contributing to the development of biocompatible materials both for diagnostic and therapeutic purposes, bioengineering of extracellular vesicles and cells derived from patients has allowed designing ad hoc systems and univocal targeting strategies. In this review, we will provide an in-depth analysis of the most innovative advances in basic and applied cancer research.
Collapse
Affiliation(s)
- Carlotta Pucci
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, 56025 Pisa, Italy
| | - Chiara Martinelli
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, 56025 Pisa, Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, 56025 Pisa, Italy.,Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy
| |
Collapse
|
34
|
Jan R, Chaudhry GES. Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics. Adv Pharm Bull 2019; 9:205-218. [PMID: 31380246 PMCID: PMC6664112 DOI: 10.15171/apb.2019.024] [Citation(s) in RCA: 504] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/16/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022] Open
Abstract
Various physiological processes involve appropriate tissue developmental process and homeostasis - the pathogenesis of several diseases connected with deregulatory apoptosis process. Apoptosis plays a crucial role in maintaining a balance between cell death and division, evasion of apoptosis results in the uncontrolled multiplication of cells leading to different diseases such as cancer. Currently, the development of apoptosis targeting anticancer drugs has gained much interest since cell death induced by apoptosis causes minimal inflammation. The understanding of complexities of apoptosis mechanism and how apoptosis is evolved by tumor cells to oppose cell death has focused research into the new strategies designed to induce apoptosis in cancer cells. This review focused on the underlying mechanism of apoptosis and the dysregulation of apoptosis modulators involved in the extrinsic and intrinsic apoptotic pathway, which include death receptors (DRs) proteins, cellular FLICE inhibitory proteins (c-FLIP), anti-apoptotic Bcl-2 proteins, inhibitors of apoptosis proteins (IAPs), tumor suppressor (p53) in cancer cells along with various current clinical approaches aimed to selectively induce apoptosis in cancer cells.
Collapse
Affiliation(s)
- Rehmat Jan
- Institute of Marine Biotechnology, Universiti Terengganu Malaysia, 21030 Terengganu, Malaysia
| | - Gul-E-Saba Chaudhry
- Institute of Marine Biotechnology, Universiti Terengganu Malaysia, 21030 Terengganu, Malaysia
| |
Collapse
|
35
|
Xiang Q, Tang J, Luo Q, Xue J, Tao Y, Jiang H, Tian J, Fan C. In vitro study of anti-ER positive breast cancer effect and mechanism of 1,2,3,4-6-pentyl-O-galloyl-beta-d-glucose (PGG). Biomed Pharmacother 2019; 111:813-820. [DOI: 10.1016/j.biopha.2018.12.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/30/2018] [Accepted: 12/14/2018] [Indexed: 01/16/2023] Open
|
36
|
Yao F, Jiang D, Cheng F, Yao W, Chen P, Guo S, Cao Y, Zhang L. Diterpene pekinenal from euphorbia pekinensis radix induced IEC-6 cells apoptosis mediated by mitochondria and death receptors. Toxicol In Vitro 2019; 57:1-8. [PMID: 30710624 DOI: 10.1016/j.tiv.2019.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/17/2019] [Accepted: 01/25/2019] [Indexed: 01/09/2023]
Abstract
Pekinenal, a diterpenoid from the roots of Euphorbia pekinensis Rupr., can cause serious intestinal toxicity. However, its toxic mechanism hasn't been comprehensively understood. This present study aims to clarify its toxic effects and investigate the potential mechanism. In vitro effects of pekinenal on cell proliferation, cell cycle and apoptosis were examined by performing experiments on rat intestinal crypt epithelial cells (IEC-6). Proteins and enzymes involved in cell apoptotic pathways were detected by Western blot and enzyme-linked immunosorbent assay (ELISA), and related mRNAs were detected by RT-PCR. The results showed that the cell cycle was arrested in G0/G1 phase, and apoptotic morphology changes in pekinenal-treated cells. Furthermore, pekinenal up-regulated the expression level of apoptotic protein including Bax, AIF, Apaf-1 and the expression level of mRNA such as Fas, FasL, TNFR1 and NF-κB, while down-regulated the expression level of Bcl-2, ultimately triggering the apoptosis of caspase dependence. In conclusion, the above data confirmed that pekinenal inhibited the proliferation of IEC-6 cells and induced cells apoptosis by modulating mitochondrial and death receptor pathways.
Collapse
Affiliation(s)
- Fang Yao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dongjing Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fangfang Cheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weifeng Yao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Peidong Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shuchen Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yudan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Li Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
37
|
Specific Cytotoxic Effects of Parasporal Crystal Proteins Isolated from Native Saudi Arabian Bacillus thuringiensis Strains against Cervical Cancer Cells. Molecules 2019; 24:molecules24030506. [PMID: 30708936 PMCID: PMC6384957 DOI: 10.3390/molecules24030506] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 11/29/2022] Open
Abstract
Currently, global efforts are being intensified towards the discovery of local Bacillus thuringiensis (Bt) isolates with unique anticancer properties. Parasporins (PS) are a group of Bt non-insecticidal crystal proteins with potential and specific in vitro anticancer activity. However, despite the significant therapeutic potential of PS-producing Bt strains, our current knowledge on the effects of these proteins is limited. Hence, the main objective of this study was to screen Bt-derived parasporal toxins for cytotoxic activities against colon (HT-29) and cervical (HeLa) cancerous cell lines. Nine non-larvicidal and non-hemolytic Bt strains, native to Saudi Arabia, were employed for the isolation of their parasporal toxins. 16S rDNA sequencing revealed a 99.5% similarity with a reference Bt strain. While PCR screening results indicated the absence of selected Cry (Cry4A, Cry4B, Cry10 and Cry11), Cyt (Cyt1 and Cyt2) and PS (PS2, PS3 and PS4) genes, it concluded presence of the PS1 gene. SDS-PAGE analysis revealed that proteolytically-cleavaged PS protein profiles exhibit patterns resembling those observed with PS1Aa1, with major bands at 56 kDa and 17 kDa (Bt7), and 41 kDa and 16 kDa (Bt5). Solubilized and trypsinized PS proteins from all Bt strains exhibited a marked and dose-dependent cytotoxicity against HeLa cancerous cells but not against HT-29 cells. IC50 values ranged from 3.2 (Bt1) to 14.2 (Bt6) with an average of 6.8 µg/mL. The observed cytotoxicity of PS proteins against HeLa cells was specific as it was not evident against normal uterus smooth muscle cells. RT-qPCR analysis revealed the overexpression of caspase 3 and caspase 9 by 3.7, and 4.2 folds, respectively, indicative of the engagement of intrinsic pathway of apoptosis. To the best of our knowledge, this is the first report exploring and exploiting the versatile repertoire of Saudi Arabian environmental niches for the isolation of native and possibly novel Saudi Bt strains with unique and specific anticancer activity. In conclusion, native Saudi Bt-derived PS proteins might have a potential to join the arsenal of natural anticancer drugs.
Collapse
|
38
|
Wu T, Dong X, Yu D, Shen Z, Yu J, Yan S. Natural product pectolinarigenin inhibits proliferation, induces apoptosis, and causes G2/M phase arrest of HCC via PI3K/AKT/mTOR/ERK signaling pathway. Onco Targets Ther 2018; 11:8633-8642. [PMID: 30584322 PMCID: PMC6284530 DOI: 10.2147/ott.s186186] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is characterized by considerable phenotypic and molecular heterogeneity, but the overall survival of HCC patients remains extremely poor. Thus, novel and efficient alternatives to antitumor agents are urgently needed. Pectolinarigenin, a flavonoid compound extract, has been previously reported for the treatment of nasopharyngeal cancer. However, the potential antitumor roles of pectolinarigenin in HCC have not been clearly elaborated. In the present study, we investigated its role in HCC treatment and explored the potential molecular mechanism(s). Materials and methods HCC cell lines SMMC7721 and PLC5 were cultured and treated with indicated concentrations of pectolinarigenin. For the HCC cell proliferation, after HCC cells were stimulated with indicated concentrations of pectolinarigenin, the cell viability was detected in CCK-8 and colony-forming assays. HCC cell invasion/migration assay was performed by Transwell and wound scratch methods. Additionally, cellular apoptosis and cell cycle arrest analysis was performed with flow cytometric analysis. Finally, the involved underlying signaling pathway, the PI3K/AKT/mTOR/ERK signaling-related molecular markers were detected through Western blot methods with indicated antibodies. Meanwhile, antitumor activity of pectolinarigenin was also assessed in tumor-bearing mice. Results The results indicated that the treatment with pectolinarigenin significantly inhibited cell proliferation and migratory and invasive abilities of SMMC7721 and PLC5 cells in concentration- and time-dependent manner. Meanwhile, pectolinarigenin markedly induced cell apoptosis and G2/M phase arrest in SMMC7721 and PLC5 cells, which was associated with apoptosis- and cell cycle-related protein levels, respectively. Furthermore, pectolinarigenin inhibited PI3K/AKT/mTOR/ERK signaling pathway. It also significantly suppressed HCC tumor growth in vivo. Conclusion Pectolinarigenin could suppress the viability and motility and cause apoptosis and G2/M phase arrest in HCC cell lines by inhibiting the PI3K/AKT/mTOR/ERK signaling pathway. This might be an appealing potential therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Tianchun Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China, .,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, People's Republic of China, .,State Key Laboratory & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China,
| | - Xiaogang Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Dongdong Yu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China, .,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, People's Republic of China, .,State Key Laboratory & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China,
| | - Zhenhua Shen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China, .,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, People's Republic of China, .,State Key Laboratory & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China,
| | - Jinbei Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Sheng Yan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China, .,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, People's Republic of China, .,State Key Laboratory & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China,
| |
Collapse
|
39
|
Xu J, Shen W, Pei B, Wang X, Sun D, Li Y, Xiu L, Liu X, Lu Y, Zhang X, Yue X. Xiao Tan He Wei Decoction reverses MNNG-induced precancerous lesions of gastric carcinoma in vivo and vitro: Regulation of apoptosis through NF-κB pathway. Biomed Pharmacother 2018; 108:95-102. [PMID: 30218863 DOI: 10.1016/j.biopha.2018.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/06/2018] [Accepted: 09/03/2018] [Indexed: 02/08/2023] Open
Abstract
In recent years, Chinese medicine has played an important role in the prognosis of gastric cancer. Precancerous lesions of gastric carcinoma (PLGC) is a class of gastric cancer which is closely related to the gastric mucosal pathology changes in the role of carcinogenic incentives, and plays key role in the progression of normal gastric mucosal cells into gastric cancerous cells. In current experiment, we explore the relationship between Chinese traditional medicine (Xiao Tan He Wei Decoction) and gastric cancer in the PLGC rat animal models and epithelial-mesenchymal transitioned GES-1 cells which were induced useing 1- Methyl-3-nitro-1-nitrosoguanidine (MNNG). PLGC rat model showed significant deterioration in the gastric mucosa with terrible growth rate in body weight and more atypical hyperplasia in gastric mucosa. MC cells, MNNG induced GES-1 cells which epithelial- mesenchymal-transition (EMT)-related proteins have a great change compare with normal GES-1 cells. The cells had characteristics of malignant cells including proliferation, invasion and metastasis ability. Our research founds that Xiao Tan He Wei Decoction could inhibit cell proliferation and increased apoptosis by increase the level of pro-apoptotic proteins like Bax and caspase-3 and decreased the level of anti-apoptotic protein Bcl-2, block the cells in G0/G1 phase simultaneously. Furthermore, Xiao Tan He Wei Decoction could inhibit nuclear factor kappa-light-chain-enhancer (NF-kB) activity and inhibit its transfer from the cytoplasm to the nucleus. However, when we incubated with NF-κB activator PMA, the effect of Xiao Tan He Wei Decoction was reversed. These results suggested that Xiao Tan He Wei Decoction could be used as a method for the treatment of gastric precancerous lesions, and possibly provide a theoretical basis for the clinical treatment of gastric cancer and gastric precancerous lesions.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Wei Shen
- Changjiang Road Community Health Service Center, NO. 639, Tonghe Road, Zhangmiao Street, Baoshan Qv, Shanghai, 200431, China
| | - Bei Pei
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Xiaowei Wang
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Dazhi Sun
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Yongjin Li
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - LiJuan Xiu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Xuan Liu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Ye Lu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Xuan Zhang
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| | - XiaoQiang Yue
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| |
Collapse
|
40
|
Abstract
Prostate cancer is the second-most widespread cancer in men worldwide. Treatment choices are limited to prostatectomy, hormonal therapy, and radiotherapy, which commonly have deleterious side effects and vary in their efficacy, depending on the stage of the disease. Among novel experimental strategies, gene therapy holds great promise for the treatment of prostate cancer. However, its use is currently limited by the lack of delivery systems able to selectively deliver the therapeutic genes to the tumors after intravenous administration without major drawbacks. To remediate this problem, a wide range of nonviral delivery approaches have been developed to specifically deliver DNA-based therapeutic agents to their site of action. This review provides an overview of the various nonviral delivery strategies and gene therapy concepts used to deliver therapeutic DNA to prostate cancer cells, and focuses on recent therapeutic advances made so far.
Collapse
Affiliation(s)
- Najla Altwaijry
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK,
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK,
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK,
| |
Collapse
|
41
|
Development of a novel cationic liposome: Evaluation of liposome mediated transfection and anti-proliferative effects of miR-101 in acute myeloid leukemia. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Lu Z, Xu H, Yu X, Wang Y, Huang L, Jin X, Sui D. 20(S)-Protopanaxadiol induces apoptosis in human hepatoblastoma HepG2 cells by downregulating the protein kinase B signaling pathway. Exp Ther Med 2018; 15:1277-1284. [PMID: 29434714 PMCID: PMC5776618 DOI: 10.3892/etm.2017.5594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/26/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatoblastoma is the most common primary liver tumor for children aged <5 years old. 20(S)-Protopanaxadiol (PPD) is a ginsenoside extracted from Pananx quinquefolium L., which inhibits tumor growth in several cancer cell lines. The purpose of the present study was to assess the anticancer activities of 20(S)-PPD in human hepatoblastoma HepG2 cells. The cytotoxicity of 20(S)-PPD on HepG2 cells was evaluated using an MTT assay. Apoptosis was detected using DAPI staining and flow cytometry. The expression of apoptosis-associated proteins was identified by western blotting. The results demonstrated that 20(S)-PPD inhibited the viability of HepG2 cell in a dose and time-dependent manner. The IC50 values were 81.35, 73.5, 48.79 µM at 24, 48 and 72 h, respectively. Topical morphological changes of apoptotic body formation following 20(S)-PPD treatment were detected by DAPI staining. The percentage of Annexin V-fluoroscein isothyiocyanate positive cells were 3.73, 17.61, 23.44 and 65.43% in HepG2 cells treated with 0, 40, 50 and 60 µM of 20(S)-PPD, respectively. Furthermore, 20(S)-PPD upregulated the expression of Bax and downregulated the expression of Bcl-2 and also activated caspases-3 and −9, and Poly [ADP-ribose] polymerase cleavage. In addition, 20(S)-PPD inhibited the phosphorylation of protein kinase B (Akt; Ser473). The results indicate that 20(S)-PPD inhibits the viability of HepG2 cells and induces apoptosis in HepG2 cells by inhibiting the phosphoinositide-3-kinase/Akt pathway.
Collapse
Affiliation(s)
- Zeyuan Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuchen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Long Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xin Jin
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
43
|
He G, Di X, Sun X, Yan J, Zhang S. Analysis of radio-sensitization patents in China from 2006 to 2015. Expert Opin Ther Pat 2017. [PMID: 28621575 DOI: 10.1080/13543776.2017.1344222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Radiotherapy is by means of ionizing radiation to kill tumor cells, inhibit and control the growth, metastasis and diffusion of tumor cells. During the last few decades, application of radiotherapy combined with chemotherapy and surgery are clinical mainstream treatments. However, little is known what radio-sensitization agents have been patented in China and what the potential drug candidates for patents are in China. Areas covered: This reviews covers research and patent literature of the last 10 years dealing with the discovery and development of novel radio-sensitization patents in China. Expert opinion: The 94 radio-sensitization patents granted from 2006 to 2015 mainly focus on six types of products. They are: traditional Chinese medicines (TCM), synthetic compounds, combinations of synthetic compounds and TCM, biological products, medical apparatus and others. In the course of tumor treatment, radiotherapy occupies an irreplaceable position. Previously believed that due to the prevalence of hypoxic cells in solid tumors, most of the tumor exist a certain degree of radiation resistance. To find effective ways to improve the sensitivity of tumor cells to radiation therapy has become a focus in scientific research and clinical treatment. So radiation sensitivity has been proposed and widely studied.
Collapse
Affiliation(s)
- Guofeng He
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Xiaoke Di
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Xinchen Sun
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Jingjing Yan
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Shu Zhang
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China.,b Clinical Research Center , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| |
Collapse
|
44
|
Apoptin Gene Delivery by the Functionalized Polyamidoamine (PAMAM) Dendrimer Modified with Ornithine Induces Cell Death of HepG2 Cells. Polymers (Basel) 2017; 9:polym9060197. [PMID: 30970874 PMCID: PMC6432117 DOI: 10.3390/polym9060197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 02/02/2023] Open
Abstract
The use of tumor-specific therapeutic agents is a promising option for efficient and safe nonviral gene transfer in gene therapy. In this study, we describe the efficacy of polyamidoamine (PAMAM)-based nonviral gene delivery carriers, namely, an ornithine conjugated PAMAM (PAMAM-O) dendrimer in delivering apoptin, a tumor-specific killer gene, into human hepatocellular carcinoma (HepG2 cells) and dermal fibroblasts. We analyzed the transfection efficiency by the luciferase assay and assessed cell viability in both cell types. The transfection efficiency of the PAMAM-O dendrimer was found to be higher than that of the PAMAM dendrimer. Moreover, the cytotoxicity of the PAMAM-O dendrimer was very low. We treated both cell types with a polyplex of PAMAM-O dendrimer with apoptin, and analyzed its cellular uptake and localization by confocal microscopy. Cell cycle distribution, tetramethylrhodamine, ethyl ester (TMRE) analysis, and transmission electron microscopy imaging showed that apoptin induced cell death in HepG2 cells. We therefore demonstrated that a PAMAM-O/apoptin polyplex can be used as an effective therapeutic strategy in cancer owing to its effectiveness as a suitable nonviral gene vector for gene therapy.
Collapse
|
45
|
El Sharkawi FZ, Ewais SM, Fahmy RH, Rashed LA. PTEN and TRAIL genes loaded zein nanoparticles as potential therapy for hepatocellular carcinoma. J Drug Target 2017; 25:513-522. [PMID: 28140697 DOI: 10.1080/1061186x.2017.1289536] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene therapy is one of the recent approaches in treatment of hepatocellular carcinoma (HCC). Development of a vector or vehicle that can selectively and efficiently deliver the gene to target cells with minimal toxicity is an urgent demand. In the present study, phosphatase and tensin homolog (PTEN) and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) genes were loaded to zein nanoparticles (ZNPs). The formulated PTEN and TRAIL-loaded ZNPs were tested for their in vitro and in vivo potential antitumor efficacy using liver tumor cells (HepG2) and HCC-induced rats as animal model. Also, mRNA expression of p53, VGEF and MMP-2 were carried out as markers of apoptosis, angiogenesis and metastasis in animal liver tissues. The results of the study showed that both PTEN and TRAIL-loaded ZNPs proved anti-proliferative activity against HepG2 cell lines with IC50 values of 0.09, 0.25 µg/ml, respectively. In vivo assay confirmed decrease in mRNA expression of both VEGF and MMP-2 with increased in P53 expression level in liver tissues of the treated animals. Therefore, authors introduced new integration between gene therapy and nanotechnology in the form of PTEN and TRAIL-loaded ZNPs that proved potential to be used in gene therapy for the treatment of HCC.
Collapse
Affiliation(s)
- Fathia Zaki El Sharkawi
- a Department of Biochemistry and Molecular Biology, Faculty of Pharmacy , Helwan University , Cairo , Egypt
| | - Shaimaa Mohammed Ewais
- a Department of Biochemistry and Molecular Biology, Faculty of Pharmacy , Helwan University , Cairo , Egypt
| | - Rania Hassan Fahmy
- b Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy , Cairo University , Cairo , Egypt.,c Department of Pharmaceutics, Faculty of Pharmacy , Ahram Canadian University , Giza , Egypt
| | - Laila Ahmed Rashed
- d Department of Biochemistry and Molecular Biology, Faculty of Medicine , Cairo University , Cairo , Egypt
| |
Collapse
|
46
|
Liu Z, Liu H, Han P, Gao F, Dahlstrom KR, Li G, Owzar K, Zevallos JP, Sturgis EM, Wei Q. Apoptotic capacity and risk of squamous cell carcinoma of the head and neck. Eur J Cancer 2017; 72:166-176. [PMID: 28033527 PMCID: PMC5287407 DOI: 10.1016/j.ejca.2016.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/28/2016] [Accepted: 11/20/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Tobacco smoke and alcohol drinking are the major risk factors for squamous cell carcinoma of the head and neck (SCCHN). Smoking and drinking cause DNA damage leading to apoptosis, and insufficient apoptotic capacity may favour development of cancer because of the dysfunction of removing damaged cells. In the present study, we investigated the association between camptothecin (CPT)-induced apoptotic capacity and risk of SCCHN in a North American population. METHODS In a case-control study of 708 SCCHN patients and 685 matched cancer-free controls, we measured apoptotic capacity in cultured peripheral blood lymphocytes in response to in vitro exposure to CPT by using the flow cytometry-based method. RESULTS We found that the mean level of apoptotic capacity in the cases (45.9 ± 23.3%) was significantly lower than that in the controls (49.0 ± 23.1%) (P = 0.002). When we used the median level of apoptotic capacity in the controls as the cutoff value for calculating adjusted odds ratios, subjects with a reduced apoptotic capacity had an increased risk (adjusted odds ratio = 1.42, 95% confidence interval = 1.13-1.78, P = 0.002), especially for those who were age ≥57 (1.73, 1.25-2.38, 0.0009), men (1.76, 1.36-2.27, <0.0001) and ever drinkers (1.67, 1.27-2.21, 0.0003), and these variables significantly interacted with apoptotic capacity (Pinteraction = 0.015, 0.005 and 0.009, respectively). A further fitted prediction model suggested that the inclusion of apoptotic capacity significantly improved in the prediction of SCCHN risk. CONCLUSION Individuals with a reduced CPT-induced apoptotic capacity may be at an increased risk of developing SCCHN, and apoptotic capacity may be a biomarker for susceptibility to SCCHN.
Collapse
Affiliation(s)
- Zhensheng Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Peng Han
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Fengqin Gao
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kristina R Dahlstrom
- Department of Head and Neck Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Guojun Li
- Department of Head and Neck Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Jose P Zevallos
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erich M Sturgis
- Department of Head and Neck Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
47
|
Bae Y, Green ES, Kim GY, Song SJ, Mun JY, Lee S, Park JI, Park JS, Ko KS, Han J, Choi JS. Dipeptide-functionalized polyamidoamine dendrimer-mediated apoptin gene delivery facilitates apoptosis of human primary glioma cells. Int J Pharm 2016; 515:186-200. [PMID: 27732896 DOI: 10.1016/j.ijpharm.2016.09.083] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 09/23/2016] [Accepted: 09/29/2016] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiform (GBM) is the most frequent and aggressive form of brain tumors in adults. However, the development of more efficient and safe nonviral vector gene therapy represents a promising therapeutic approach, using a tumor-specific killer gene, named apoptin. In this study, we describe the efficacy of non-viral gene delivery vectors, the amino acid-conjugated PAMAM derivatives (PAMAM-H-R and PAMAM-H-K) in delivering a therapeutic gene, displaying affinity toward human primary glioma cells (GBL-14 cells) and dermal fibroblasts. We analyzed transfection efficiency, using luciferase (Luci) and a pDNA encoding for enhanced fluorescent protein (EGFP), and cytotoxicity in both cells. The results show that transfection efficiency of PAMAM-H-R improved compared to native PAMAM dendrimer, but cytotoxicity of PAMAM-H-R and PAMAM-H-K were very low. We treated both cells with a polyplex formation of PAMAM-H-R or PAMAM-H-K/apoptin, and analyzed their cellular uptake and localization by flow cytometry and confocal microscopy. Furthermore, we analyzed the endosomal escape effect using TEM images, and found that PAMAM-H-R showed very fast escape from endosome to the cytosol. Caspase 3 activity assay, cell cycle distribution, and JC-1 analysis showed apoptosis induced by apoptin in GBL-14 cells. This indicates that PAMAM-H-R can be a potential nonviral vector gene delivery carrier for brain tumor therapy. The present study demonstrates that PAMAM-H-R/apoptin gene polyplex can be used as an effective therapeutic candidate for GBM due to its selective induction of apoptosis in primary glioma cells as a potential nonviral gene delivery carrier for brain tumor therapy.
Collapse
Affiliation(s)
- Yoonhee Bae
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 614-735, Republic of Korea
| | - Eric S Green
- Salt Lake Community College, Salt Lake City, UT, USA
| | - Goo-Young Kim
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Su Jeong Song
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Ji Young Mun
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Republic of Korea
| | - Sunray Lee
- Cell Engineering for Origin Research Center 46-21, Susong-dog, Jongno-gu, Seoul 110-140, Republic of Korea
| | - Jong-Il Park
- Chungnam National University School of Medicine, Daejeon 301-747, Republic of Korea
| | - Jong-Sang Park
- School of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Kyung Soo Ko
- Department of Internal Medicine, Sanggye Paik Hospital, Cardiovascular and Metabolic Disease Center, Inje University, Seoul 139-707, Republic of Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 614-735, Republic of Korea.
| | - Joon Sig Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Republic of Korea.
| |
Collapse
|
48
|
Kim H, Shin EA, Kim CG, Lee DY, Kim B, Baek NI, Kim SH. Obovatol Induces Apoptosis in Non-small Cell Lung Cancer Cells via C/EBP Homologous Protein Activation. Phytother Res 2016; 30:1841-1847. [PMID: 27489231 DOI: 10.1002/ptr.5690] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/03/2016] [Accepted: 07/11/2016] [Indexed: 12/27/2022]
Abstract
Although obovatol, a phenolic compound from the bark of Magnolia obovata, was known to have antioxidant, neuroprotective, antiinflammatory, antithrombotic and antitumour effects, its underlying antitumour mechanism is poorly understood so far. Thus, in the present study, the antitumour molecular mechanism of obovatol was investigated in non-small cell lung cancer cells (NSCLCs). Obovatol exerted cytotoxicity in A549 and H460 NSCLCs, but not in BEAS-2B cells. Also, obovatol increased sub-G1 accumulation and early and late apoptotic portion in A549 and H460 NSCLCs. Consistently, obovatol cleaved PARP, activated caspase 9/3 and Bax and attenuated the expression of cyclin D1 in A549 and H460 NSCLCs. Interestingly, obovatol upregulated the expression of endoplasmic reticulum stress proteins such as C/EBP homologous protein (CHOP), IRE1α, ATF4 and p-elF2 in A549 and H460 NSCLCs. Conversely, depletion of CHOP blocked the apoptotic activity of obovatol to increase sub-G1 accumulation in A549 and H460 NSCLCs. Overall, our findings support scientific evidences that obovatol induces apoptosis via CHOP activation in A549 and H460 NSCLCs. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Heejeong Kim
- Department of East West Medical Science, Graduate School of East West Medical Science, Kyung Hee University, Yongin, 446-701, Korea
| | - Eun Ah Shin
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea
| | - Chang Geun Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration (RDA), Eumseong, 27709, Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea
| | - Nam-In Baek
- Department of Oriental Medicine Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 131-701, Korea.
| |
Collapse
|
49
|
Ma K, Zhang C, Huang MY, Li WY, Hu GQ. Cinobufagin induces autophagy-mediated cell death in human osteosarcoma U2OS cells through the ROS/JNK/p38 signaling pathway. Oncol Rep 2016; 36:90-8. [PMID: 27176794 PMCID: PMC4899018 DOI: 10.3892/or.2016.4782] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
The main objective of this study was to explore whether autophagy could be triggered by cinobufagin, and to clarify the role of autophagy in the antitumor effects of cinobufagin on U2OS cells and the underlying mechanisms. U2OS cells were exposed to 15, 30, 60 and 120 mg/l cinobufagin for 0, 12, 24 and 48 h. An MTT assay was used to measure cell viability. FITC-Annexin Ⅴ/PI staining and flow cytometry were used to analyze the apoptotic ratio, while apoptotic morphological changes were assessed by PI and Hoechst 33258 viable cell staining. The effects of autophagy on the cells were investigated with GFP-LC3b green fluorescence plasmid transfection and transmission electron microscopy. The levels of caspase-3, -8, - 9, cleaved PARP, LC3-II/LC3-I, p62 and the activation of JNK/p-38 were detected by western blot analysis. Reactive oxygen species (ROS) fluorescence intensity was examined under fluorescence microscopy with an analysis software system. Cell proliferation was obviously inhibited by cinobufagin in a dose- and time-dependent manner. The apoptosis ratio was gradually increased with treatment time as evidenced by flow cytometric analysis and Hoechst 33258 staining. Exposure to cinobufagin resulted in the activation of caspase-3, -8, -9, as well as cleaved PARP which indicated that cinobufagin induced caspase-dependent apoptosis. Autophagy was confirmed in the cinobufagin-treated cells as evidenced by formation of autophagosomes, accumulation of GFP-LC3 fluorescence particles as well as the upregulation of LC3-II/LC3-I levels. Inhibition of autophagy diminished apoptosis as detected by the MTT assays. Moreover the percentage of apoptotic cells decreased following pretreatment with 3-MA, CQ and si-beclin-1. Cinobufagin also induced phosphorylation of the JNK and p38 signaling pathway as well as ROS generation. The JNK and p38 inhibitors significantly attenuated coexistence of apoptosis and autophagy-related proteins. The ROS scavenger also prevented phosphorylation of the JNK and p38 signaling pathway. Our research proved that cinobufagin triggered apoptosis and autophagic cell death via activation of the ROS/JNK/p-38 axis.
Collapse
Affiliation(s)
- Kun Ma
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Chuan Zhang
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Man-Yu Huang
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Wu-Yin Li
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Guo-Qiang Hu
- College of Pharmacy, Henan University, Kaifeng, Henan 475000, P.R. China
| |
Collapse
|
50
|
Kim J, Yun M, Kim E, Jung D, Won G, Kim B, Jung JH, Kim S. Decursin enhances TRAIL-induced apoptosis through oxidative stress mediated- endoplasmic reticulum stress signalling in non-small cell lung cancers. Br J Pharmacol 2016; 173:1033-44. [PMID: 26661339 PMCID: PMC5341238 DOI: 10.1111/bph.13408] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/25/2015] [Accepted: 12/03/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE The TNF-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent due to its remarkable ability to selectively kill tumour cells. However, because most tumours exhibit resistance to TRAIL-induced apoptosis, the development of combination therapies to overcome resistance to TRAIL is required for effective cancer therapy. EXPERIMENTAL APPROACH Cell viability and possible synergy between the plant pyranocoumarin decursin and TRAIL was measured by MTT assay and calcusyn software. Reactive oxygen species (ROS) and apoptosis were measured using dichlorodihydrofluorescein and annexin/propidium iodide in cell flow cytometry. Changes in protein levels were assessed with Western blotting. KEY RESULTS Combining decursin and TRAIL markedly decreased cell viability and increased apoptosis in TRAIL-resistant non-small-cell lung cancer (NSCLC) cell lines. Decursin induced expression of the death receptor 5 (DR5). Inhibition of DR5 attenuated apoptotic cell death in decursin + TRAIL treated NSCLC cell lines. Interestingly, induction of DR5 and CCAAT/enhancer-binding protein homologues protein by decursin was mediated through selective induction of the pancreatic endoplasmic reticulum kinase (PERK)/activating transcription factor 4 (ATF4) branch of the endoplasmic reticulum stress response pathway. Furthermore, enhancement of PERK/ATF4 signalling by decursin was mediated by ROS generation in NSCLC cell lines, but not in normal human lung cells. Decursin also markedly down-regulated expression of survivin and Bcl-xL in TRAIL-resistant NSCLC cells. CONCLUSIONS AND IMPLICATIONS ROS generation by decursin selectively activated the PERK/ATF4 axis of the endoplasmic reticulum stress signalling pathway, leading to enhanced TRAIL sensitivity in TRAIL-resistant NSCLC cell lines, partly via up-regulation of DR5.
Collapse
Affiliation(s)
- Jaekwang Kim
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Miyong Yun
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Eun‐Ok Kim
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Deok‐Beom Jung
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Gunho Won
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Bonglee Kim
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Ji Hoon Jung
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| | - Sung‐Hoon Kim
- College of Korean MedicineKyung Hee UniversitySeoulSouth Korea
| |
Collapse
|