1
|
Manoharan S, Murugesan K, Gunasekaran S, Vedagiri H, Perumal E. Quinazoline-2,4(1H,3H)-dione modulates STAT3 and FOXO3a signaling in HepG2 cells. Bioorg Chem 2025; 157:108304. [PMID: 40022846 DOI: 10.1016/j.bioorg.2025.108304] [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: 12/03/2024] [Revised: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Hepatocellular carcinoma (HCC), the most prominent type of primary liver cancer, often diagnosed late, leading to poor prognosis and limited treatment options. This study investigated the anti-carcinogenic effect of Quinazoline-2,4(1H,3H)-dione (Qd), a quinazoline derivative of natural origin and identified Qd as an effective compound against HCC via STAT3 and FOXO3a signaling. STAT3 and FOXO3a are two well-known molecular drivers of HCC. In silico findings revealed Qd as the potent candidate due to its highly stable interaction with STAT3 and FOXO3a. To validate its anticancer activity, in vitro experiments were conducted on the HepG2 cell line. Qd exerts cytotoxic effect in HepG2 cells with an IC50 value of 26.07 μM, while being non-toxic in WRL-68 cells at a lower concentrations with an IC50 of 326.5 μM. Morphological changes and apoptotic cell death were confirmed using DAPI staining and Live/Dead assay. Qd also induced ROS-mediated mitochondrial damage. Qd upregulated mRNA expressions of pro-apoptotic and necroptotic markers while downregulating anti-apoptotic marker. Accordingly, the protein expression analysis demonstrated increased levels of Bax, Caspase 3, c-PARP, RIPK1, RIPK3 and MLKL, while decreasing Bcl2 and PARP expressions. Gene and protein expression of STAT3 remained at a basal level while FOXO3a gene expression increased significantly at 5 μM Qd concentration. Significant changes were particularly observed at 5 μM Qd concentration in all in vitro experiments. Despite quinazoline compounds have been shown biological and pharmacological effects, the anticancer effect of Qd is elusive till date. These in silico and in vitro findings highlighted Qd as a potent compound for further exploration in HCC therapy by targeting apoptotic and necroptotic cell death pathways.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore - 641046, India
| | - Krishnasanthiya Murugesan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore - 641046, India
| | - Sinduja Gunasekaran
- Molecular Genomics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore - 641046, India
| | - Hemamalini Vedagiri
- Molecular Genomics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore - 641046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore - 641046, India.
| |
Collapse
|
2
|
Cho WK, Choi HJ, Ma JY. Antiviral Activity of Berbamine Against Influenza A Virus Infection. Int J Mol Sci 2025; 26:2819. [PMID: 40141460 PMCID: PMC11942913 DOI: 10.3390/ijms26062819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Berbamine (BBM) is a bibenzyl isoquinoline present in the traditional Chinese herbal medicine Berberis amurensisis Rupr. The present study demonstrates that BBM exerts strong antiviral efficacy against influenza A virus (IAV) infection. We examined the anti-IAV effect of BBM using green fluorescent protein (GFP)-expressing influenza A and H1N1 IAV. The fluorescence microscopy, fluorescence-activated cell sorting analysis, and plaque assay showed that BBM significantly hinders IAV infection. The immunofluorescence analysis confirmed the anti-influenza activity of BBM. From the time-of-addition and hemagglutination inhibition results, it is elucidated that the antiviral effect of BBM is closely related to its inhibitory effect against viral binding and entry at an early infection stage. Our findings imply that BBM has the potential to be developed as a potent antiviral drug against influenza viral infection.
Collapse
Affiliation(s)
- Won-Kyung Cho
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, 70 Chemdanro, Dong-gu, Daegu 41062, Republic of Korea;
| | | | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, 70 Chemdanro, Dong-gu, Daegu 41062, Republic of Korea;
| |
Collapse
|
3
|
Li L, Lv M, Li Y, Sun H, Li J, Li W, Wang X, Bi R, Zhang Z, Bo Z, Shen H, Wang J, Zhuansun M, Zhou J, Xue Y, Suo X, Tong R, Sun P. Berbamine inhibits Pseudorabies virus in vitro and in vivo. Vet Microbiol 2025; 301:110356. [PMID: 39733660 DOI: 10.1016/j.vetmic.2024.110356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Pseudorabies virus (PRV) is a significant pathogen that causes acute infectious diseases in pigs, resulting in considerable economic losses for the global pig industry. The lack of effective control measures and vaccines against the circulating variants of PRV highlights the pressing need for novel treatment strategies. In this study, a screening of a natural product library identified Berbamine as a promising compound that inhibits PRV replication, with a selectivity index of 17. Preliminary investigations demonstrated that Berbamine impedes viral proliferation by targeting the replication and release stages of the PRV life cycle. In experiments with mice artificially infected with PRV, Berbamine significantly alleviated clinical symptoms and histopathological changes in brain tissue caused by PRV infection. Furthermore, molecular docking studies indicated that Berbamine targets the UL50 protein, not only of PRV but also of HSV-1, FHV-1, and BoHV-1. Given that the UL50 protein is a promising target for antiviral drug development, Berbamine holds considerable potential for broad application in antiviral therapies.
Collapse
Affiliation(s)
- Liang Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
| | - Muze Lv
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yangfan Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Huihui Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jie Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wenyan Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xuan Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ruimin Bi
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zuyao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zongyi Bo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Haixiao Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jun Wang
- Anhui Provincial Center for Animal Disease Control and Prevention, Hefei, Anhui 230091, China
| | - Minghao Zhuansun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jinchi Zhou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yuting Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xinru Suo
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Tong
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Pei Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
4
|
Xiang H, Qiao J, Lin H, Li J, Li Y, Sun H, Wang X, Bi R, Zhang Z, Bo Z, Shen H, Zhou J, Tong R, Suo X, Xue Y, Li L, Sun P. Berbamine inhibits porcine epidemic diarrhea virus in vitro and in vivo. Vet Microbiol 2024; 298:110244. [PMID: 39236425 DOI: 10.1016/j.vetmic.2024.110244] [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: 07/11/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a significant contributor to high mortality rates in piglets, posing a serious threat to the global pig industry. The absence of effective control measures and vaccines against circulating PEDV variants underscores the urgent need for new treatment strategies. In this study, we screened a compound library and identified Berbamine as a potential anti-PEDV drug through molecular docking techniques. In vitro experiments demonstrated that Berbamine significantly inhibits PEDV proliferation in Vero and IPEC-J2 cells in a dose-dependent manner, primarily targeting the replication phase of the PEDV life cycle. Furthermore, in vivo experiments revealed that Berbamine effectively alleviates intestinal damage caused by PEDV infection in piglets, leading to a reduction in viral load and cytokine levels, including IL-6, IL-8, IL-1β, and TNF-α. Additionally, autodock predictions indicate that viral non-structural proteins 3 and 16 (Nsp3 and Nsp16) are potential targets for Berbamine. Consequently, Berbamine holds significant promise for application and development as an antiviral treatment against PEDV.
Collapse
Affiliation(s)
- Hongwei Xiang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jixue Qiao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Haicheng Lin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jie Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yangfan Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Huihui Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xuan Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ruimin Bi
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zuyao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zongyi Bo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Haixiao Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinchi Zhou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Tong
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xinru Suo
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yuting Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Liang Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China.
| | - Pei Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
5
|
Peng H, He Y, Hu Y, Sheng S, Maitiyasen M, Li J, Liu Y, Hou X, Song H, Yi J. Berbamine promotes ferroptosis of esophageal squamous cell carcinoma by facilitating USP51-mediated GPX4 ubiquitination and degradation. Biomed Pharmacother 2024; 179:117309. [PMID: 39151312 DOI: 10.1016/j.biopha.2024.117309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
Esophageal cancer ranks among the most prevalent malignant tumors globally. The prognosis for esophageal squamous cell carcinoma remains poor, with a 5-year survival rate below 20 % due to limited advances in therapy. Ferroptosis, a novel form of iron-dependent lipid peroxidation-driven regulated cell death (RCD), shows significant promise in cancer treatment. Berbamine (BBM), a natural bisbenzylisoquinoline alkaloid derived from Berberis amurensis, exhibits anti-tumor effects against various cancers, yet its impact on esophageal cancer remains to be elucidated. This study aimed to explore the role of BBM in inducing ferroptosis in the treatment of esophageal cancer, focusing on its molecular mechanisms. Gene set enrichment analysis(GSEA) analysis highlighted the potential of BBM as an anti-cancer agent through ferroptosis induction. We found that BBM inhibited growth and epithelial-mesenchymal transition (EMT) in esophageal cancer cell lines, promoting Fe accumulation, ROS, and malondialdehyde (MDA) production, thereby triggering cell death. These suppressive effects were successfully reversed by Ferrostatin-1 (Fer-1). Mechanistically, BBM decreased deubiquitination enzyme USP51 levels, leading to ubiquitin degradation and glutathione peroxidase 4(GPX4) instability, and it stimulated ferroptosis. The Overexpression of USP51 mitigated the downregulation of GPX4 induced by BBM.BBM significantly inhibited tumor xenograft growth in nude mice. This discovery positions BBM as a promising therapeutic candidate for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Hao Peng
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - YuanPeng He
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuepeng Hu
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Siqi Sheng
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Maierhaba Maitiyasen
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jingfeng Li
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yvxuan Liu
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyu Hou
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haizhu Song
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Jun Yi
- Department of Cardiothoracic Surgery,Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
6
|
Qureshi SS, Nizamuddin S, Xu J, Vancov T, Chen C. Cellulose nanocrystals from agriculture and forestry biomass: synthesis methods, characterization and industrial applications. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:58745-58778. [PMID: 39340607 PMCID: PMC11513767 DOI: 10.1007/s11356-024-35127-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Agricultural and forestry biomass wastes, often discarded or burned without adequate management, lead to significant environmental harm. However, cellulose nanocrystals (CNCs), derived from such biomass, have emerged as highly promising materials due to their unique properties, including high tensile strength, large surface area, biocompatibility, and renewability. This review provides a detailed analysis of the lignocellulosic composition, as well as the elemental and proximate analysis of different biomass sources. These assessments help determine the yield and characteristics of CNCs. Detailed discussion of CNC synthesis methods -ranging from biomass pretreatment to hydrolysis techniques such as acid, mineral, solid acid, ionic liquid, and enzymatic methods-are provided. The key physical, chemical, and thermal properties of CNCs are also highlighted, particularly in relation to their industrial applications. Recommendations for future research emphasize the need to optimize CNC synthesis processes, identify suitable biomass feedstocks, and explore new industrial applications.
Collapse
Affiliation(s)
- Sundus Saeed Qureshi
- Australian Rivers Institute and School of Environment and Science, Griffith University, Nathan Campus, Brisbane, Queensland, 4111, Australia
- Cooperative Research Centre for High Performance Soils, Callaghan, NSW, Australia
| | - Sabzoi Nizamuddin
- Water Regulation Division, Grampians Wimmera Mallee Water (GWMWater) Corporation, Horsham, Victoria, 3400, Australia
| | - Jia Xu
- Australian Rivers Institute and School of Environment and Science, Griffith University, Nathan Campus, Brisbane, Queensland, 4111, Australia
- Cooperative Research Centre for High Performance Soils, Callaghan, NSW, Australia
| | - Tony Vancov
- NSW Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, NSW, 2568, Australia
| | - Chengrong Chen
- Australian Rivers Institute and School of Environment and Science, Griffith University, Nathan Campus, Brisbane, Queensland, 4111, Australia.
- Cooperative Research Centre for High Performance Soils, Callaghan, NSW, Australia.
| |
Collapse
|
7
|
Bhutkar M, Saha A, Tomar S. Viral methyltransferase inhibitors: berbamine, venetoclax, and ponatinib as efficacious antivirals against chikungunya virus. Arch Biochem Biophys 2024; 759:110111. [PMID: 39111614 DOI: 10.1016/j.abb.2024.110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/20/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
Chikungunya virus (CHIKV), transmitted by mosquitoes, poses a significant global health threat. Presently, no effective treatment options are available to reduce the disease burden. The lack of approved therapeutics against CHIKV and the complex spectrum of chronic musculoskeletal and neurological manifestations raise significant concerns, and repurposing drugs could offer swift avenues in the development of effective treatment strategies. RNA capping is a crucial step meditated by non-structural protein 1 (nsP1) in CHIKV replication. In this study, FDA-approved antivirals targeting CHIKV nsP1 methyltransferase (MTase) have been identified by structure-based virtual screening. Berbamine Hydrochloride (BH), ABT199/Venetoclax (ABT), and Ponatinib (PT) were the top-hits, which exhibited robust binding energies. Tryptophan fluorescence spectroscopy-based assay confirmed binding of BH-, ABT-, and PT to purified nsP1 with KD values ∼5.45 μM, ∼161.3 μM, and ∼3.83 μM, respectively. In a capillary electrophoresis-based assay, a decrease in CHIKV nsP1 MTase activity was observed in a dose-dependent manner. Treatment with BH, ABT, and PT lead to a dose-dependent reduction in the virus titer with IC50 < 100, ∼6.75, and <3.9 nM, respectively, and reduced viral mRNA levels. The nsP1 MTases are highly conserved among alphaviruses; therefore, BH, ABT, and PT, as expected, inhibited replication machinery in Sindbis virus (SINV) replicon assay with IC50 ∼1.94, ∼0.23, and >1.25 μM, respectively. These results highlight the potential of repurposing drugs as rapid and effective antiviral therapeutics against CHIKV.
Collapse
Affiliation(s)
- Mandar Bhutkar
- Molecular Virology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| | - Ankita Saha
- Molecular Virology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| | - Shailly Tomar
- Molecular Virology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India.
| |
Collapse
|
8
|
Takayanagi SI, Chuganji S, Tanaka M, Wang B, Hasegawa S, Fukumoto K, Wasano N, Kakitani M, Ochiai N, Kawai Y, Ueda T, Ishikawa A, Kurimoto Y, Fukui A, Kamibayashi S, Imai E, Kunisato A, Nozawa H, Kaneko S. A culture method with berbamine, a plant alkaloid, enhances CAR-T cell efficacy through modulating cellular metabolism. Commun Biol 2024; 7:685. [PMID: 38834758 PMCID: PMC11150386 DOI: 10.1038/s42003-024-06297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Memory T cells demonstrate superior in vivo persistence and antitumor efficacy. However, methods for manufacturing less differentiated T cells are not yet well-established. Here, we show that producing chimeric antigen receptor (CAR)-T cells using berbamine (BBM), a natural compound found in the Chinese herbal medicine Berberis amurensis, enhances the antitumor efficacy of CAR-T cells. BBM is identified through cell-based screening of chemical compounds using induced pluripotent stem cell-derived T cells, leading to improved viability with a memory T cell phenotype. Transcriptomics and metabolomics using stem cell memory T cells reveal that BBM broadly enhances lipid metabolism. Furthermore, the addition of BBM downregulates the phosphorylation of p38 mitogen-activated protein kinase and enhanced mitochondrial respiration. CD19-CAR-T cells cultured with BBM also extend the survival of leukaemia mouse models due to their superior in vivo persistence. This technology offers a straightforward approach to enhancing the antitumor efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Shin-Ichiro Takayanagi
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan.
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
- Biomedical Science Research Laboratories 2, Research Division, Kyowa Kirin Co., Ltd., Tokyo, Japan.
| | - Sayaka Chuganji
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Tanaka
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Bo Wang
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Saki Hasegawa
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ken Fukumoto
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Nariaki Wasano
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
| | - Makoto Kakitani
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
| | - Nakaba Ochiai
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yohei Kawai
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tatsuki Ueda
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akihiro Ishikawa
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuko Kurimoto
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
| | - Asami Fukui
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
| | - Sanae Kamibayashi
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eri Imai
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Kunisato
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
| | - Hajime Nozawa
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa, Kanagawa, 251-8555, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
9
|
Manoharan S, Saha S, Murugesan K, Santhakumar A, Perumal E. Natural bioactive compounds and STAT3 against hepatocellular carcinoma: An update. Life Sci 2024; 337:122351. [PMID: 38103726 DOI: 10.1016/j.lfs.2023.122351] [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: 09/17/2023] [Revised: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is a challenging and very fatal liver cancer. The signal transducer and activator of transcription 3 (STAT3) pathway is a crucial regulator of tumor development and are ubiquitously active in HCC. Therefore, targeting STAT3 has emerged as a promising approach for preventing and treating HCC. Various natural bioactive compounds (NBCs) have been proven to target STAT3 and have the potential to prevent and treat HCC as STAT3 inhibitors. Numerous kinds of STAT3 inhibitors have been identified, including small molecule inhibitors, peptide inhibitors, and oligonucleotide inhibitors. Due to the undesirable side effects of the conventional therapeutic drugs against HCC, the focus is shifted to NBCs derived from plants and other natural sources. NBCs can be broadly classified into the categories of terpenes, alkaloids, carotenoids, and phenols. Most of the compounds belong to the family of terpenes, which prevent tumorigenesis by inhibiting STAT3 nuclear translocation. Further, through STAT3 inhibition, terpenes downregulate matrix metalloprotease 2 (MMP2), matrix metalloprotease 9 (MMP9) and vascular endothelial growth factor (VEGF), modulating metastasis. Terpenes also suppress the anti-apoptotic proteins and cell cycle markers. This review provides comprehensive information related to STAT3 abrogation by NBCs in HCC with in vitro and in vivo evidences.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Shreejit Saha
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Krishnasanthiya Murugesan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Aksayakeerthana Santhakumar
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India.
| |
Collapse
|
10
|
Almeida-Pinto F, Pinto R, Rocha J. Navigating the Complex Landscape of Ebola Infection Treatment: A Review of Emerging Pharmacological Approaches. Infect Dis Ther 2024; 13:21-55. [PMID: 38240994 PMCID: PMC10828234 DOI: 10.1007/s40121-023-00913-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
In 1976 Ebola revealed itself to the world, marking the beginning of a series of localized outbreaks. However, it was the Ebola outbreak that began in 2013 that incited fear and anxiety around the globe. Since then, our comprehension of the virus has been steadily expanding. Ebola virus (EBOV), belonging to the Orthoebolavirus genus of the Filoviridae family, possesses a non-segmented, negative single-stranded RNA genome comprising seven genes that encode multiple proteins. These proteins collectively orchestrate the intricate process of infecting host cells. It is not possible to view each protein as monofunctional. Instead, they synergistically contribute to the pathogenicity of the virus. Understanding this multifaceted replication cycle is crucial for the development of effective antiviral strategies. Currently, two antibody-based therapeutics have received approval for treating Ebola virus disease (EVD). In 2022, the first evidence-based clinical practice guideline dedicated to specific therapies for EVD was published. Although notable progress has been made in recent years, deaths still occur. Consequently, there is an urgent need to enhance the therapeutic options available to improve the outcomes of the disease. Emerging therapeutics can target viral proteins as direct-acting antivirals or host factors as host-directed antivirals. They both have advantages and disadvantages. One way to bypass some disadvantages is to repurpose already approved drugs for non-EVD indications to treat EVD. This review offers detailed insight into the role of each viral protein in the replication cycle of the virus, as understanding how the virus interacts with host cells is critical to understanding how emerging therapeutics exert their activity. Using this knowledge, this review delves into the intricate mechanisms of action of current and emerging therapeutics.
Collapse
Affiliation(s)
| | - Rui Pinto
- Faculdade de Farmácia, Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines (iMED.ULisboa), 1649-003, Lisbon, Portugal
- Dr. Joaquim Chaves, Medicine Laboratory, Joaquim Chaves Saúde (JCS), Carnaxide, Portugal
| | - João Rocha
- Faculdade de Farmácia, Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines (iMED.ULisboa), 1649-003, Lisbon, Portugal
| |
Collapse
|
11
|
Hossen MS, Hasan MN, Haque M, Al Arian T, Halder SK, Uddin MJ, Abdullah-Al-Mamun M, Shakil MS. Immunoinformatics-aided rational design of multiepitope-based peptide vaccine (MEBV) targeting human parainfluenza virus 3 (HPIV-3) stable proteins. J Genet Eng Biotechnol 2023; 21:162. [PMID: 38055114 DOI: 10.1186/s43141-023-00623-5] [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: 07/14/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Human parainfluenza viruses (HPIVs) are common RNA viruses responsible for respiratory tract infections. Human parainfluenza virus 3 (HPIV-3) is particularly pathogenic, causing severe illnesses with no effective vaccine or therapy available. RESULTS The current study employed a systematic immunoinformatic/reverse vaccinology approach to design a multiple epitope-based peptide vaccine against HPIV-3 by analyzing the virus proteome. On the basis of a number of therapeutic features, all three stable and antigenic proteins with greater immunological relevance, namely matrix protein, hemagglutinin neuraminidase, and RNA-directed RNA polymerase L, were chosen for predicting and screening suitable T-cell and B-cell epitopes. All of our desired epitopes exhibited no homology with human proteins, greater population coverage (99.26%), and high conservancy among reported HPIV-3 isolates worldwide. All of the T- and B-cell epitopes are then joined by putative ligands, yielding a 478-amino acid-long final construct. Upon computational refinement, validation, and thorough screening, several programs rated our peptide vaccine as biophysically stable, antigenic, allergenic, and non-toxic in humans. The vaccine protein demonstrated sufficiently stable interaction as well as binding affinity with innate immune receptors TLR3, TLR4, and TLR8. Furthermore, codon optimization and virtual cloning of the vaccine sequence in a pET32a ( +) vector showed that it can be readily expressed in the bacterial system. CONCLUSION The in silico designed HPIV-3 vaccine demonstrated potential in evoking an effective immune response. This study paves the way for further preclinical and clinical evaluation of the vaccine, offering hope for a future solution to combat HPIV-3 infections.
Collapse
Affiliation(s)
- Md Sakib Hossen
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka, 1213, Bangladesh.
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216, Bangladesh.
| | - Md Nazmul Hasan
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216, Bangladesh.
- Department of Biochemistry and Molecular Biology, Hajee Mohammad Danesh Science and Technology University, Dinajpur, 5200, Bangladesh.
| | - Munima Haque
- Biotechnology Program, Department of Mathematics and Natural Sciences (MNS), Brac University, kha-208, 1 Bir Uttam Rafiqul Islam Ave, Dhaka, 1212, Bangladesh
| | - Tawsif Al Arian
- Department of Pharmacy, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Sajal Kumar Halder
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md Jasim Uddin
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - M Abdullah-Al-Mamun
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md Salman Shakil
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216, Bangladesh
- Microbiology Program, Department of Mathematics and Natural Sciences (MNS), Brac University, 66 Mohakhali, Dhaka, 1212, Bangladesh
| |
Collapse
|
12
|
Chen X, Zhang L, He L, Zheng L, Tuo B. Potassium channels as novel molecular targets in hepatocellular carcinoma (Review). Oncol Rep 2023; 50:185. [PMID: 37654193 PMCID: PMC10485806 DOI: 10.3892/or.2023.8622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Hepatocellular carcinoma (HCC) poses a serious health burden worldwide. It is often not diagnosed until the patient is at an advanced stage of the disease, when treatment options are limited and the prognosis is poor. Therefore, novel treatment strategies are urgently required. Potassium (K+) channels have an important role in HCC, including regulating the proliferation, migration, invasion and drug resistance of HCC cells. The aim of the present review was therefore to survey the relevant publications that have investigated K+ channels not only as markers for the early diagnosis of HCC, but also as potential therapeutic targets for the treatment of HCC. Several of these channels have been indicated to be the sites of action for natural products previously known to inhibit HCC; however, more systematic studies are required to determine which K+ channels may be utilized for the clinical treatment of HCC, particularly in the advanced stages of the disease and in cases where patients are resistant to the existing drugs.
Collapse
Affiliation(s)
- Xingyue Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ling He
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Liming Zheng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
13
|
Wu X, Zhang D, Qiao X, Zhang L, Cai X, Ji J, Ma JA, Zhao Y, Belperio JA, Boström KI, Yao Y. Regulating the cell shift of endothelial cell-like myofibroblasts in pulmonary fibrosis. Eur Respir J 2023; 61:2201799. [PMID: 36758986 PMCID: PMC10249020 DOI: 10.1183/13993003.01799-2022] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023]
Abstract
Pulmonary fibrosis is a common and severe fibrotic lung disease with high morbidity and mortality. Recent studies have reported a large number of unwanted myofibroblasts appearing in pulmonary fibrosis, and shown that the sustained activation of myofibroblasts is essential for unremitting interstitial fibrogenesis. However, the origin of these myofibroblasts remains poorly understood. Here, we create new mouse models of pulmonary fibrosis and identify a previously unknown population of endothelial cell (EC)-like myofibroblasts in normal lung tissue. We show that these EC-like myofibroblasts significantly contribute myofibroblasts to pulmonary fibrosis, which is confirmed by single-cell RNA sequencing of human pulmonary fibrosis. Using the transcriptional profiles, we identified a small molecule that redirects the differentiation of EC-like myofibroblasts and reduces pulmonary fibrosis in our mouse models. Our study reveals the mechanistic underpinnings of the differentiation of EC-like myofibroblasts in pulmonary fibrosis and may provide new strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- These authors contributed equally to this work
| | - Daoqin Zhang
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- These authors contributed equally to this work
| | - Xiaojing Qiao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaden Ji
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jocelyn A Ma
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yan Zhao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John A Belperio
- Division of Pulmonary and Critical Care Medicine, Clinical Immunology, and Allergy, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- The Molecular Biology Institute at UCLA, Los Angeles, CA, USA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
14
|
Tuli HS, Rath P, Chauhan A, Ramniwas S, Vashishth K, Varol M, Jaswal VS, Haque S, Sak K. Phloretin, as a Potent Anticancer Compound: From Chemistry to Cellular Interactions. Molecules 2022; 27:8819. [PMID: 36557950 PMCID: PMC9787340 DOI: 10.3390/molecules27248819] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Phloretin is a natural dihydrochalcone found in many fruits and vegetables, especially in apple tree leaves and the Manchurian apricots, exhibiting several therapeutic properties, such as antioxidant, antidiabetic, anti-inflammatory, and antitumor activities. In this review article, the diverse aspects of the anticancer potential of phloretin are addressed, presenting its antiproliferative, proapoptotic, antimetastatic, and antiangiogenic activities in many different preclinical cancer models. The fact that phloretin is a planar lipophilic polyphenol and, thus, a membrane-disrupting Pan-Assay Interference compound (PAIN) compromises the validity of the cell-based anticancer activities. Phloretin significantly reduces membrane dipole potential and, therefore, is expected to be able to activate a number of cellular signaling pathways in a non-specific way. In this way, the effects of this minor flavonoid on Bax and Bcl-2 proteins, caspases and MMPs, cytokines, and inflammatory enzymes are all analyzed in the current review. Moreover, besides the anticancer activities exerted by phloretin alone, its co-effects with conventional anticancer drugs are also under discussion. Therefore, this review presents a thorough overview of the preclinical anticancer potential of phloretin, allowing one to take the next steps in the development of novel drug candidates and move on to clinical trials.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida 201303, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India
| | - Seema Ramniwas
- University Centre for Research & Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali 140413, India
| | - Kanupriya Vashishth
- Advance Cardiac Centre Department of Cardiology, Post Graduate Institute of Medical Education and Research (PGIMER) Chandigarh, Chandigarh 160012, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla 48000, Turkey
| | - Vivek Sheel Jaswal
- Department of Chemistry and Chemical Science, School of Physical & Material Sciences, Central University of Himachal Pradesh, Dharamshala 176206, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
15
|
Roy NS, Park NI, Kim NS, Park Y, Kim BY, Kim YD, Yu JK, Kim YI, Um T, Kim S, Choi IY. Comparative Transcriptomics for Genes Related to Berberine and Berbamine Biosynthesis in Berberidaceae. PLANTS (BASEL, SWITZERLAND) 2022; 11:2676. [PMID: 36297700 PMCID: PMC9610958 DOI: 10.3390/plants11202676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Berberine and berbamine are bioactive compounds of benzylisoquinoline alkaloids (BIAs) present in Berberis species. The contents of berbamine are 20 times higher than berberine in leaf tissues in three closely related species: Berberis koreana, B. thunbergii and B. amurensis. This is the first report on the quantification of berberine compared to the berbamine in the Berberis species. Comparative transcriptome analyses were carried out with mRNAs from the leaf tissues of the three-species. The comparison of the transcriptomes of B. thunbergii and B. amurensis to those of B. koreana, B. thunbergii showed a consistently higher number of differentially expressed genes than B. amurensis in KEGG and DEG analyses. All genes encoding enzymes involved in berberine synthesis were identified and their expressions were variable among the three species. There was a single copy of CYP80A/berbamunine synthase in B. koreana. Methyltransferases and cytochrome P450 mono-oxidases (CYPs) are key enzymes for BIA biosynthesis. The current report contains the copy numbers and other genomic characteristics of the methyltransferases and CYPs in Berberis species. Thus, the contents of the current research are valuable for molecular characterization for the medicinal utilization of the Berberis species.
Collapse
Affiliation(s)
- Neha Samir Roy
- Agriculture and Life Sciences Research Institute, Kangwon National University, Chuncheon 24341, Korea
| | - Nam-Il Park
- Department of Plant Science, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Nam-Soo Kim
- NBIT, Kangwon National University, Gangwondaehakgil-1, Bodeumkwan 504, Chuncheon 24341, Korea
| | - Yeri Park
- Department of Plant Science, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Bo-Yun Kim
- Plant Resources Division, National Institute of Biological Resources, Incheon 22689, Korea
| | - Young-Dong Kim
- Department of Life Science, Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Korea
| | - Ju-Kyung Yu
- Syngenta Crop Protection LLC, 9 Davis Drive, Research Triangle Park, NC 27709, USA
| | - Yong-In Kim
- On Biological Resource Research Institute, Chuncheon 24239, Korea
| | - Taeyoung Um
- Agriculture and Life Sciences Research Institute, Kangwon National University, Chuncheon 24341, Korea
| | - Soonok Kim
- Microorganism Resources Division, National Institute of Biological Resources, Incheon 22689, Korea
| | - Ik-Young Choi
- Agriculture and Life Sciences Research Institute, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
16
|
Zheng Z, Zhang L, Hou X. Potential roles and molecular mechanisms of phytochemicals against cancer. Food Funct 2022; 13:9208-9225. [PMID: 36047380 DOI: 10.1039/d2fo01663j] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increasing evidence has been reported regarding phytochemicals, plant secondary metabolites, having therapeutic functions against numerous human diseases. Recently, phytochemicals (flavonoids, polyphenols, terpenoids, alkaloids, saponins, coumarins and so on) have shown promising anti-cancer efficacy with their distinct advantages of high efficiency and low toxicity. They regulate programmed cell death (apoptosis, pyroptosis, and autophagy), migration and senescence-related signaling pathways of cancer via the modulation of reactive oxygen species (ROS), mitogen activated protein kinase (MAPK) pathway, deleted in liver cancer 1 (DLC1), nuclear factor κ light-chain-enhancer of activated B cell (NF-κB) pathways and glycolytic enzymes. Here, we review the molecular mechanisms by which phytochemicals prevent the development of cancer. Furthermore, phytochemicals combined with chemotherapeutic agents could target the crosstalk among multiple signal cascades to block chemoresistance and attenuate carcinogenic properties, and can be considered as a novel and potential therapeutic strategy. Our review highlights that the mechanisms and promising applications are required to be understood to decisively establish the anti-cancer efficacy of natural phytochemicals.
Collapse
Affiliation(s)
- Zhaodi Zheng
- School of Forensics and Laboratory Medicine, Jining Medical University, Jining, 272067, Shandong, China.
| | - Leilei Zhang
- School of Forensics and Laboratory Medicine, Jining Medical University, Jining, 272067, Shandong, China.
| | - Xitan Hou
- School of Forensics and Laboratory Medicine, Jining Medical University, Jining, 272067, Shandong, China.
| |
Collapse
|
17
|
Li H, Luo K, Yang Z, Chen M, Yang X, Wang J, Ying Y, Wu D, Wang Q. Berbamine Suppresses the Growth of Gastric Cancer Cells by Inactivating the BRD4/c-MYC Signaling Pathway. Drug Des Devel Ther 2022; 16:129-141. [PMID: 35046638 PMCID: PMC8762520 DOI: 10.2147/dddt.s338881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Berbamine (Ber), a bioactive constituent extracted from a traditional Chinese medicinal herb, has been shown to exhibit broad inhibitory activity on a panel of cancer cell types. However, its effects and the underlying molecular mechanisms on gastric cancer (GC) remain poorly understood. METHODS The anti-growth activity of Ber on two GC cell lines and normal gastric epithelial cell line were evaluated using MTS and clone formation assay. Flow cytometry analysis was employed to evaluate the cell cycle distribution and apoptosis of GC cells. Western blot and quantitative PCR (qPCR) analysis were employed to investigate the anti-GC mechanism of Ber. The inhibitory activity and binding affinity of Ber against BRD4 were evaluated by homogeneous time-resolved fluorescence (HTRF) and surface plasmon resonance (SPR) assay, respectively. Molecular docking and molecular simulations were conducted to predict the interaction mode between BRD4 and Ber. RESULTS The results demonstrated that Ber reduced the proliferation of GC cell lines SGC-7901 and BGC-823 and induced cell cycle arrest and apoptosis. Mechanistically, Ber was identified as a novel natural-derived BRD4 inhibitor through multiple experimental assay, and its anti-GC activity was probably mediated by BRD4 inhibition. Molecular modeling studies suggested that Ber might bind to BRD4 primarily through hydrophobic interactions. CONCLUSION Our study uncovered the underlying anti-GC activity of Ber in vitro and suggested that Ber holds promise as a potential lead compound in the discovery of novel BRD4 inhibitors.
Collapse
Affiliation(s)
- Hongchun Li
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Kexue Luo
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Zhuying Yang
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Miao Chen
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, People's Republic of China
| | - Xiuyun Yang
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Jiesheng Wang
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, People's Republic of China
| | - Dengxuan Wu
- Department of Rehabilitation Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Qinxian Wang
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, People's Republic of China
| |
Collapse
|
18
|
Ko D, Kim E, Shin EA, Nam SH, Yoon J, Lee JS, Lee Y, Park S, Ha K, Choi SY, Lee JW, Kim S. Therapeutic effects of TM4SF5-targeting chimeric and humanized monoclonal antibodies in hepatocellular and colon cancer models. Mol Ther Oncolytics 2022; 24:452-466. [PMID: 35211652 PMCID: PMC8841960 DOI: 10.1016/j.omto.2022.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022] Open
Abstract
The transmembrane 4 L six family member 5 (TM4SF5) is aberrantly expressed in hepatocellular and colorectal cancers, and has been implicated in tumor progression, suggesting that it could serve as a novel therapeutic target. Previously, we screened a murine antibody phage-display library to generate a novel monoclonal antibody, Ab27, that is specific to the extracellular loop 2 of TM4SF5. In this study, we evaluated the effects of chimeric Ab27 using cancer cells expressing endogenous TM4SF5 or stably overexpressing TM4SF5 in vivo and in vitro. Monotherapy with Ab27 significantly decreased tumor growth in liver and colon cancer xenograft models, including a sorafenib-resistant model, and decreased the phosphorylation of focal adhesion kinase (FAK), p27Kip1, and signal transducer and activator of transcription 3 (STAT3). No general Ab27 toxicity was observed in vivo. Combination treatment with Ab27 and sorafenib or doxorubicin exerted higher antitumor activity than monotherapy. In addition, we humanized the Ab27 sequence by the complementarity-determining region (CDR) grafting method. The humanized antibody Ab27-hz9 had reduced immunogenicity but exhibited target recognition and antitumor activity comparable with those of Ab27. Both Ab27 and Ab27-hz9 efficiently targeted tumor cells expressing TM4SF5 in vivo. These observations strongly support the further development of Ab27-hz9 as a novel therapeutic agent against liver and colorectal cancers.
Collapse
Affiliation(s)
- Dongjoon Ko
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejon 34113, Korea
| | - Eunmi Kim
- Department of Pharmacy Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul 08826, Korea
| | - Eun-Ae Shin
- Department of Pharmacy Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul 08826, Korea
| | - Seo Hee Nam
- Department of Pharmacy Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul 08826, Korea
| | - Junghwa Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Korea
| | - Jin-Sook Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Korea
| | - Yunhee Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Korea
| | - Sora Park
- Antibody Drug Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Korea
| | - Kyungsoo Ha
- Drug Efficacy Evaluation Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Korea
| | - So-Young Choi
- Protein Drug Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Korea
| | - Jung Weon Lee
- Department of Pharmacy Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-Gu, Seoul 08826, Korea
| | - Semi Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejon 34113, Korea
| |
Collapse
|
19
|
Farooqi AA, Wen R, Attar R, Taverna S, Butt G, Xu B. Regulation of Cell-Signaling Pathways by Berbamine in Different Cancers. Int J Mol Sci 2022; 23:ijms23052758. [PMID: 35269900 PMCID: PMC8911410 DOI: 10.3390/ijms23052758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 02/26/2022] [Indexed: 12/18/2022] Open
Abstract
Natural product research is a cornerstone of the architectural framework of clinical medicine. Berbamine is a natural, potent, pharmacologically active biomolecule isolated from Berberis amurensis. Berbamine has been shown to modulate different oncogenic cell-signaling pathways in different cancers. In this review, we comprehensively analyze how berbamine modulates deregulated pathways (JAK/STAT, CAMKII/c-Myc) in various cancers. We systematically analyze how berbamine induces activation of the TGF/SMAD pathway for the effective inhibition of cancer progression. We also summarize different nanotechnological strategies currently being used for proficient delivery of berbamine to the target sites. Berbamine has also been reported to demonstrate potent anti-cancer and anti-metastatic effects in tumor-bearing mice. The regulation of non-coding RNAs by berbamine is insufficiently studied, and future studies must converge on the identification of target non-coding RNAs. A better understanding of the regulatory role of berbamine in the modulation of non-coding RNAs and cell-signaling pathways will be advantageous in the effective translation of laboratory findings to clinically effective therapeutics.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan;
| | - Ru Wen
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA;
| | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University, Istanbul 34755, Turkey;
| | - Simona Taverna
- Institute for Biomedical Research and Innovation, National Research Council of Italy, 90146 Palermo, Italy;
- Institute of Translational Pharmacology (IFT-CNR), National Research Council of Italy, 90146 Palermo, Italy
| | - Ghazala Butt
- Institute of Botany, University of the Punjab, Lahore 54590, Pakistan;
| | - Baojun Xu
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai 519087, China
- Correspondence: ; Tel.: +86-756-2620636
| |
Collapse
|
20
|
Nguyen VK, Kou KGM. The biology and total syntheses of bisbenzylisoquinoline alkaloids. Org Biomol Chem 2021; 19:7535-7543. [PMID: 34524341 DOI: 10.1039/d1ob00812a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This mini-review provides a concise overview of the biosynthetic pathway and pharmacology of bisbenzylisoquinoline alkaloid (bisBIA) natural products. Additional emphasis is given to the methodologies in the total syntheses of both simpler acyclic diaryl ether dimers and their macrocyclic counterparts bearing two diaryl ether linkages.
Collapse
Affiliation(s)
- Viviene K Nguyen
- Department of Chemistry, University of California, Riverside, California 92521, USA.
| | - Kevin G M Kou
- Department of Chemistry, University of California, Riverside, California 92521, USA.
| |
Collapse
|
21
|
Yang S, Yang S, Zhang H, Hua H, Kong Q, Wang J, Jiang Y. Targeting Na + /K + -ATPase by berbamine and ouabain synergizes with sorafenib to inhibit hepatocellular carcinoma. Br J Pharmacol 2021; 178:4389-4407. [PMID: 34233013 DOI: 10.1111/bph.15616] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The multikinase inhibitor sorafenib is a first-line drug for advanced hepatocellular carcinoma. The response to sorafenib varies among hepatocellular carcinoma patients and many of the responders suffer from reduced sensitivity after long-term treatment. This study aims to explore a novel strategy to potentiate or maximize the anti-hepatocellular carcinoma effects of sorafenib. EXPERIMENTAL APPROACH We used hepatocellular carcinoma cell lines, western blotting, various antagonists, siRNA and tumour xenografts mouse model to determine the anti- hepatocellular carcinoma effects of sorafenib in combination with berbamine or other Na+ /K+ -ATPase ligands. KEY RESULTS Berbamine and the cardiotonic steroid, ouabain, synergize with sorafenib to inhibit hepatocellular carcinoma cells growth. Mechanistically, berbamine induces Src phosphorylation in Na+ /K+ -ATPase-dependent manner, leading to the activation of p38MAPK and EGFR-ERK pathways. The Na+ /K+ -ATPase ligand ouabain also induces Src, EGFR, type I insulin-like growth factor receptor, ERK1/2 and p38MAPK phosphorylation in hepatocellular carcinoma cells. Treatment of hepatocellular carcinoma cells with Src or EGFR inhibitor inhibits the induction of ERK1/2 phosphorylation by berbamine. Moreover, sorafenib inhibits the induction of Src, p38MAPK, EGFR and ERK1/2 phosphorylation by berbamine and ouabain. Importantly, combination of sorafenib with berbamine or ouabain synergistically inhibits both sorafenib-naïve and sorafenib-resistant hepatocellular carcinoma cells growth. Co-treatment of hepatocellular carcinoma cells with berbamine and sorafenib significantly induces cell death and significantly inhibits hepatocellular carcinoma xenografts growth in vivo. CONCLUSION AND IMPLICATIONS Berbamine or other Na+ /K+ -ATPase ligands have a potential for improving sorafenib responsiveness in hepatocellular carcinoma. Targeting Na+ /K+ -ATPase represents a novel strategy to potentiate the anti- hepatocellular carcinoma effects of sorafenib.
Collapse
Affiliation(s)
- Songpeng Yang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shu Yang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, China
| | - Qingbin Kong
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|