1
|
Ye H, Wang Y, Zhang X, Yang L, Cai B, Zhang D, Peng B. Characterization of global research trends and prospects on celastrol, a principal bioactive ingredient of Tripterygium wilfordii Hook F: bibliometric analysis. PHARMACEUTICAL BIOLOGY 2025; 63:15-26. [PMID: 39745069 DOI: 10.1080/13880209.2024.2443424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
CONTEXT Celastrol, acknowledged as a prominent exemplar of the potential for transforming traditional medicinal compounds into contemporary pharmaceuticals, has garnered considerable attention owing to its extensive pharmacological activities. The increasing volume of publications concerning celastrol highlights its importance in current scientific inquiry. Despite the growing interest in this compound, a bibliometric analysis focused on this subject remains to be undertaken. OBJECTIVE Our study explored a bibliometric approach to identify and characterize global research trends and frontiers related to celastrol, including mapping research outputs, influential contributors, and thematic areas, as well as highlighting gaps and opportunities for future investigations. MATERIALS AND METHODS In this study, we utilized the Web of Science Core Collection (WoSCC) to source and review articles related to celastrol published from 1997 to 2023. The bibliometric analysis was conducted using the R package 'Bibliometrix,' supplemented by visualization tools including CiteSpace, VOSviewer, and GraphPad Prism 10. RESULTS Celastrol related research papers have exhibited an upward trend annually and can be categorized into three distinct phases, each highlighting different areas of focus. China, the United States, and South Korea rank as the top three nations for publication volume, with varied research interests across these countries. Several prolific research teams have emerged, each with distinct areas of interest. Examining the primary research domains of celastrol (anti-inflammatory, anticancer, and toxicity) reveals a notable intersection between the first two domains. DISCUSSION AND CONCLUSIONS The scope and depth of celastrol research have been steadily expanding, with regional and team-specific variations. Key research areas include anti-inflammatory, anticancer, and toxicity studies. Future research is expected to focus on enhancing the effectiveness and reducing the toxicity of celastrol. Meanwhile, given the multi-target characteristics of celastrol's effects, integrating methods such as network biology and molecular simulation will provide a novel perspective for celastrol research.
Collapse
Affiliation(s)
- Huizi Ye
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Yufang Wang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Xue Zhang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lin Yang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Banglan Cai
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Denghai Zhang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Bin Peng
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| |
Collapse
|
2
|
Gu T, Raval R, Bashkin Z, Zhou C, Ko S, Kong N, Hong S, Bhaskara A, Shah S, Joshi A, Thellakal S, Rim K, Marimuthu A, Venkatesan S, Wang E, Li S, Jayabalan A, Tao A, Fang Y, Xia L, Chui A, Shu E, Zhang T, Chen Z, Njoo E. Synthesis, antiproliferative activity, and biological profiling of C-19 trityl and silyl ether andrographolide analogs in colon cancer and breast cancer cells. Bioorg Med Chem Lett 2025; 121:130163. [PMID: 40043819 DOI: 10.1016/j.bmcl.2025.130163] [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: 10/14/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Andrographolide, a labdane diterpenoid isolated from Andrographis paniculata, putatively functions through covalent inhibition of NF-κB, a transcription factor that modulates tumor survival and metastasis. Previous studies have found that functionalization of the C-19 hydroxyl alters the primary mode of action from inhibition of NF-κB to the modulation of the Wnt1/β-catenin signaling pathway. Here, we synthesized a series of twelve C-19 trityl and silyl ether analogs, including three novel substituted trityl analogs and four novel substituted silyl analogs of andrographolide. MTT assays revealed cell line selectivity between colorectal and breast cancer cells, which is consistent with known mechanisms of β-catenin-driven cell proliferation in colorectal cancer cell lines. Most compounds exhibited cell line specific antiproliferative activity in HCT-116 and HT-29 colorectal cancer cell lines. Specifically, within 24 h, C-19 analogs of andrographolide exhibit far more limited antiproliferative activity in MCF-7 breast cancer cells compared to HCT-116, HT-29, and MDA-MB-231 cells. Through in vitro TNF-α-dependent NF-κB reporter and Wnt1-dependent luciferase reporter assays, we observed that several analogs generally exhibit greater inhibitory activity compared to andrographolide. Fluorescence imaging demonstrated that cells treated with andrographolide and its C-19 analogs retained similar distributions of active β-catenin, but notable differences in antiproliferative potency upon co-delivery with GSK-3β inhibitor CHIR99021 indicate that several lead compounds exhibit attenuated biological activity selectively in HT-29 cells. Collectively, this work indicates that modest structural modifications at C-19 of andrographolide can have profound implications for its biological activity in mechanisms connected to its anticancer activity.
Collapse
Affiliation(s)
- Tiffany Gu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Rushika Raval
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Zachary Bashkin
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Carina Zhou
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Sanghyuk Ko
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Natalie Kong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Seoyeon Hong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aditya Bhaskara
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samarth Shah
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Aditi Joshi
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samahith Thellakal
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Kaitlyn Rim
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Anushree Marimuthu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Srishti Venkatesan
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Emma Wang
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Sophia Li
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Aditi Jayabalan
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Alice Tao
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Yilin Fang
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Lorelei Xia
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aidan Chui
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Emily Shu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Tracy Zhang
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Zhan Chen
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Edward Njoo
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA.
| |
Collapse
|
3
|
Edet P, Ekpenyong M, Asitok A, Ubi D, Echa C, Edeghor U, Antai S. Bio-induced overproduction of heterocycloanthracin-like bacteriocin in Lysinibacillus macroides by Aspergillus austroafricanus: optimization of medium conditions and evaluation of potential applications. BMC Biotechnol 2025; 25:41. [PMID: 40405132 PMCID: PMC12101020 DOI: 10.1186/s12896-025-00979-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 05/12/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Plants and microorganisms are at the forefront of natural exploitable bioresources for the discovery of novel bioactive compounds (BACs) to provide solutions to food and agricultural challenges. The present study aimed to produce a novel biotechnologically-relevant BAC from a mangrove sediment bacterium under optimized bioprocess medium conditions. The BAC-producing bacteria were isolated via the crowded plate technique, and medium optimization was performed via sequential statistics of response surface methodology (RSM). The RSM model predictions were optimized, validated, and scaled up in a 5-L bioreactor via submerged batch fermentation. The BAC was extracted with ethyl acetate, purified via silica gel column chromatography, and identified via semipreparative high-performance liquid chromatography using bioactive standards with known retention times. The biocontrol, antioxidant and biopreservation potential of the BAC were evaluated via standard methods. RESULTS The results revealed that strain GKRMS-A9 produced the largest inhibition zone diameter (ZND) of 17 mm against the susceptible mould. The bacterium and its susceptible mould were identified as Lysinibacillus macroides and Aspergillus austroafricanus strains, respectively. Bioprocess medium optimization produced 9.6 g L- 1 of the BAC with a ZND of 47.1 mm using 44.84% [v v- 1] rice processing effluent, 8.58 gL- 1 casamino acid, 1.39 g L- 1 MgSO4.7H2O, 2.78 g L- 1 CaCl2.2H2O, 16.94% [v v- 1] inoculum volume, and 10.45 g L- 1 Na2HPO4/NaH2PO4. The BAC concentration increased 48.7-fold in response to biological induction with susceptible mould. Silica gel chromatography revealed 9 bioactive fractions in the ethyl acetate extract, with fraction C (retention time of 9.02 min) eliciting the largest mean ZND of 38.1 ± 1.7 mm against Aspergillus austroafricanus. Fraction C was identified as a heterocycloanthracin-like class II bacteriocin with a molecular weight of 10.5 kDa. CONCLUSION The bacteriocin 'macroidin' is stable over a wide range of pH values and temperatures and has significant antimicrobial activity against Gram-positive food-borne and phytopathogenic strains of bacteria and moulds. Its antioxidant activities against DPPH and ABTS*+ radicals are comparable to those of ascorbic acid, making this biomolecule a promising agent for biopreservation and phytopathogen control applications in the food and agricultural sectors.
Collapse
Affiliation(s)
- Philomena Edet
- Environmental Microbiology and Biotechnology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Maurice Ekpenyong
- Environmental Microbiology and Biotechnology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria.
- University of Calabar Collection of Microorganisms, University of Calabar, Calabar, Nigeria.
- Food and Industrial Microbiology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria.
| | - Atim Asitok
- Environmental Microbiology and Biotechnology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
- University of Calabar Collection of Microorganisms, University of Calabar, Calabar, Nigeria
| | - David Ubi
- Environmental Microbiology and Biotechnology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Cecilia Echa
- Environmental Microbiology and Biotechnology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Uwamere Edeghor
- Food and Industrial Microbiology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Sylvester Antai
- Environmental Microbiology and Biotechnology Unit, Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria.
- University of Calabar Collection of Microorganisms, University of Calabar, Calabar, Nigeria.
| |
Collapse
|
4
|
Liao GQ, Tang HM, Yu YD, Fu LZ, Li SJ, Zhu MX. Mass spectrometry-based metabolomic as a powerful tool to unravel the component and mechanism in TCM. Chin Med 2025; 20:62. [PMID: 40355943 PMCID: PMC12067679 DOI: 10.1186/s13020-025-01112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Mass spectrometry (MS)-based metabolomics has emerged as a transformative tool to unraveling components and their mechanisms in traditional Chinese medicine (TCM). The integration of advanced analytical platforms, such as LC-MS and GC-MS, coupled with metabolomics, has propelled the qualitative and quantitative characterization of TCM's complex components. This review comprehensively examines the applications of MS-based metabolomics in elucidating TCM efficacy, spanning chemical composition analysis, molecular target identification, mechanism-of-action studies, and syndrome differentiation. Recent innovations in functional metabolomics, spatial metabolomics, single-cell metabolomics, and metabolic flux analysis have further expanded TCM research horizons. Artificial intelligence (AI) and bioinformatics integration offer promising avenues for overcoming analytical bottlenecks, enhancing database standardization, and driving interdisciplinary breakthroughs. However, challenges remain, including the need for improved data processing standardization, database expansion, and understanding of metabolite-gene-protein interactions. By addressing these gaps, metabolomics can bridge traditional practices and modern biomedical research, fostering global acceptance of TCM. This review highlights the synergy of advanced MS techniques, computational tools, and TCM's holistic philosophy, presenting a forward-looking perspective on its clinical translation and internationalization.
Collapse
Affiliation(s)
- Guang-Qin Liao
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
| | - Hong-Mei Tang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- National Animal Disease-Chongqing Monitoring Station, Chongqing, 402460, China
| | - Yuan-Di Yu
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
- National Animal Disease-Chongqing Monitoring Station, Chongqing, 402460, China
| | - Li-Zhi Fu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- Chongqing Research Center of Veterinary Biologicals Engineering and Technology, Chongqing, 402460, China
| | - Shuang-Jiao Li
- Chinese Academy of Agricultural Sciences, Beijing, 100061, China
| | - Mai-Xun Zhu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China.
| |
Collapse
|
5
|
Babineau N, Dien Nguyen LT, Mathieu D, McCue C, Schlecht N, Abrahamson T, Hamberger B, Busta L. A molecular representation system with a common reference frame for analyzing triterpenoid structural diversity. PLANT COMMUNICATIONS 2025; 6:101320. [PMID: 40134219 DOI: 10.1016/j.xplc.2025.101320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/26/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Researchers have uncovered hundreds of thousands of natural products, many of which contribute to medicine, materials, and agriculture. However, missing knowledge about the biosynthetic pathways of these products hinders their expanded use. Nucleotide sequencing is key to pathway elucidation efforts, and analyses of the molecular structures of natural products, although seldom discussed explicitly, also play an important role by suggesting hypothetical pathways for testing. Structural analyses are also important in drug discovery, for which many molecular representation systems-methods of representing molecular structures in a computer-friendly format-have been developed. Unfortunately, pathway elucidation investigations seldom use these representation systems. This gap likely occurs because those systems are primarily built to document molecular connectivity and topology rather than the absolute positions of bonds and atoms in a common reference frame, which would enable chemical structures to be connected with potential underlying biosynthetic steps. Here, we expand on recently developed skeleton-based molecular representation systems by implementing a common-reference-frame-oriented system. We tested this system using triterpenoid structures as a case study and explored its applications in biosynthesis and structural diversity tasks. The common-reference-frame system can identify structural regions of high or low variability on the scale of atoms and bonds and enable hierarchical clustering that is closely connected to underlying biosynthesis. Combined with information on phylogenetic distribution, the system illuminates distinct sources of structural variability, such as different enzyme families operating in the same pathway. These characteristics outline the potential of common-reference-frame molecular representation systems to support large-scale pathway elucidation efforts.
Collapse
Affiliation(s)
- Nicole Babineau
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Le Thanh Dien Nguyen
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Davis Mathieu
- Genetics and Genome Science Graduate Program, Michigan State University, East Lansing, MI 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Clint McCue
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Nicholas Schlecht
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI 48824, USA
| | - Taylor Abrahamson
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Björn Hamberger
- Genetics and Genome Science Graduate Program, Michigan State University, East Lansing, MI 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI 48824, USA
| | - Lucas Busta
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN 55812, USA.
| |
Collapse
|
6
|
Chang KT, Chen YC, Lien Y, Huang YH, Huang CY. Inhibition of RPA32 and Cytotoxic Effects of the Carnivorous Plant Sarracenia purpurea Root Extract in Non-Small-Cell Lung Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2025; 14:1426. [PMID: 40430991 PMCID: PMC12115182 DOI: 10.3390/plants14101426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/30/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
The carnivorous plant Sarracenia purpurea has been traditionally used in various ethnobotanical applications, including treatments for type 2 diabetes and tuberculosis-like symptoms. This study investigates the cytotoxic effects of S. purpurea root extract (Sp-R) on human non-small-cell lung cancer (NSCLC) cell lines, including H1975, H838, and A549, focusing on its impact on cell survival, apoptosis, proliferation, and migration. Additionally, its ability to inhibit the single-stranded DNA-binding activity of human RPA32 (huRPA32), a key protein in DNA replication, was evaluated. Extracts from different plant parts (leaf, stem, and root) were prepared using various solvents (water, methanol, ethanol, and acetone) and screened for apoptosis-inducing potential using the chromatin condensation assay. Among these, the acetone-extracted root fraction (Sp-R-A) exhibited the most potent pro-apoptotic effects. The MTT assay demonstrated a dose-dependent cytotoxic effect on NSCLC cells, with IC50 values of 33.74 μg/mL for H1975, 60.79 μg/mL for H838, and 66.52 μg/mL for A549. Migration and clonogenic assays further revealed that Sp-R-A significantly inhibited cancer cell migration and colony formation in a dose-dependent manner. Moreover, Sp-R-A enhanced apoptosis when combined with the EGFR inhibitor afatinib, suggesting a potential synergistic effect. The electrophoretic mobility shift assay confirmed that Sp-R-A significantly inhibited the DNA-binding activity of huRPA32, with an IC50 of 13.6 μg/mL. AlphaFold structural prediction and molecular docking studies indicated that major bioactive compounds in S. purpurea, including α-amyrin, ursolic acid, and betulinaldehyde, strongly interact with the DNA-binding domain of huRPA32, potentially contributing to its inhibitory effect. Overall, these findings suggest that huRPA32 is a potential molecular target of Sp-R-A and the anticancer potential of S. purpurea root extract against NSCLC is highlighted, supporting further investigation into its therapeutic applications.
Collapse
Affiliation(s)
- Kuo-Ting Chang
- Division of Translational Medicine, Department of Research and Development, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan
| | - Yu-Cheng Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan
| | - Yi Lien
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Yen-Hua Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 402, Taiwan
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| |
Collapse
|
7
|
Rice AJ, Sword TT, Chengan K, Mitchell DA, Mouncey NJ, Moore SJ, Bailey CB. Cell-free synthetic biology for natural product biosynthesis and discovery. Chem Soc Rev 2025; 54:4314-4352. [PMID: 40104998 PMCID: PMC11920963 DOI: 10.1039/d4cs01198h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Indexed: 03/20/2025]
Abstract
Natural products have applications as biopharmaceuticals, agrochemicals, and other high-value chemicals. However, there are challenges in isolating natural products from their native producers (e.g. bacteria, fungi, plants). In many cases, synthetic chemistry or heterologous expression must be used to access these important molecules. The biosynthetic machinery to generate these compounds is found within biosynthetic gene clusters, primarily consisting of the enzymes that biosynthesise a range of natural product classes (including, but not limited to ribosomal and nonribosomal peptides, polyketides, and terpenoids). Cell-free synthetic biology has emerged in recent years as a bottom-up technology applied towards both prototyping pathways and producing molecules. Recently, it has been applied to natural products, both to characterise biosynthetic pathways and produce new metabolites. This review discusses the core biochemistry of cell-free synthetic biology applied to metabolite production and critiques its advantages and disadvantages compared to whole cell and/or chemical production routes. Specifically, we review the advances in cell-free biosynthesis of ribosomal peptides, analyse the rapid prototyping of natural product biosynthetic enzymes and pathways, highlight advances in novel antimicrobial discovery, and discuss the rising use of cell-free technologies in industrial biotechnology and synthetic biology.
Collapse
Affiliation(s)
- Andrew J Rice
- Department of Biochemistry, School of Medicine - Basic Sciences, Vanderbilt University Medical Research Building-IV, Nashville, Tennessee, 37232, USA
| | - Tien T Sword
- Department of Chemistry, University of Tennessee-Knoxville, Knoxville, TN, USA
| | | | - Douglas A Mitchell
- Department of Biochemistry, School of Medicine - Basic Sciences, Vanderbilt University Medical Research Building-IV, Nashville, Tennessee, 37232, USA
- Department of Chemistry, Vanderbilt University, Medical Research Building-IV, Nashville, Tennessee, 37232, USA
| | - Nigel J Mouncey
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, 94720, USA
| | - Simon J Moore
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - Constance B Bailey
- School of Chemistry, University of Sydney, Camperdown, NSW, 2001, Australia.
| |
Collapse
|
8
|
Sang CY, Liu JR, Zheng YD, Chai T, Shi JT, Naghavi MR, Alibekovna KE, Solievich BA, Yang JL. Obacunone potentiated PD-1 immunotherapy in pancreatic cancer by mediating CD36. Eur J Pharmacol 2025; 994:177367. [PMID: 39986594 DOI: 10.1016/j.ejphar.2025.177367] [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: 06/10/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Chemotherapy for patients with pancreatic cancer typically has a poor prognosis. Immunotherapy is currently a hot therapeutical approach to treat tumors. Various studies have shown that natural products have numerous activities, especially in the anti-tumor field. The triterpenoid class compound Obacunone has been shown to have various bioactivities, including anti-cancer properties. In this study, combining Obacunone with anti-PD-1 to treat pancreatic cancer in mice enhanced the anti-cancer activity of anti-PD-1 and suppressed tumor growth significantly. Proteomic analysis, immunofluorescence, Western blot, and flow cytometry revealed that this combination of compounds modulated the CD36-mediated PPAR signaling pathway to improve the infiltration and number of immune-associated CD4+ and CD8+ T cells in tumors. This report provides a new strategy for discovering immunotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Chun-Yan Sang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, 730000, China
| | - Jia-Rong Liu
- Department of Medicine, Northwest Minzu University, Lanzhou, 730030, China
| | - Yi-Dan Zheng
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, 730000, China
| | - Tian Chai
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, 730000, China
| | - Jiao-Tai Shi
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, 730000, China
| | | | - Komila Eshbakova Alibekovna
- S.Yu.Yunusov Institute of the Chemistry of Plant Substances, Academy of Sciences, Tashkent, 100170, Uzbekistan
| | | | - Jun-Li Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, 730000, China.
| |
Collapse
|
9
|
Chen M, Hao BC, Zhu XH, Zhang LK, Zheng YY, Zhou XJ, Schäberle TF, Shen L, Wang CY, Liu Y. Molecular networking reveals indole diterpenoids from the marine-derived fungus Penicillium sp. N4-3. MARINE LIFE SCIENCE & TECHNOLOGY 2025; 7:302-312. [PMID: 40417251 PMCID: PMC12102448 DOI: 10.1007/s42995-024-00274-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 11/29/2024] [Indexed: 05/27/2025]
Abstract
MS/MS-based molecular networking is an effective strategy to rapidly dereplicate known compounds and to guide the discovery process for new and novel natural products. In the present study, the chemical diversity of indole diterpenoids from the marine-derived fungus Penicillium sp. N4-3 was investigated using molecular networking techniques. Guided by this information, targeted isolation resulted in two new indole diterpenoids shearinines R and S (1, 2) and an oxidative artifact shearinine T (3), together with the verification of two known analogs (4, 5). Furthermore, five indole diterpenoids (6-10), including three putatively new ones, shearinines U-W (6, 9, 10), were predicted from the molecular ion cluster by the combination of GNPS molecular networking and manual analysis of MS/MS fragmentation clusters. Shearinines T (3) and W (10) are characterized by an oxidative cleavage of the C-2-C-18 double bond. Feature fragment ions of these shearinines revealed two type of dominant ions related to the indole moiety and the breaking of C-9 side chain or Ring I. Compound 1 showed antibacterial activities against a panel of pathogenic bacteria with IC50 values ranging from 6.34 to 47.96 μg/mL and inhibited the growth of the human hepatic (HepG2) and gastric (SGC-7901) cancer cells lines with IC50 values of 6.27 and 19.16 μg/mL, respectively. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-024-00274-6.
Collapse
Affiliation(s)
- Min Chen
- Marine Science & Technology Institute, College of Environmental Science & Engineering, Yangzhou University, 196#, Huayang West Street, Yangzhou, 225127 China
| | - Bao-Cong Hao
- Marine Science & Technology Institute, College of Environmental Science & Engineering, Yangzhou University, 196#, Huayang West Street, Yangzhou, 225127 China
| | - Xia-Hao Zhu
- Marine Science & Technology Institute, College of Environmental Science & Engineering, Yangzhou University, 196#, Huayang West Street, Yangzhou, 225127 China
| | - Li-Kui Zhang
- Marine Science & Technology Institute, College of Environmental Science & Engineering, Yangzhou University, 196#, Huayang West Street, Yangzhou, 225127 China
| | - Yao-Yao Zheng
- Key Laboratory of Marine Drugs, The Ministry of Education of China, Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237 China
| | - Xiao-Jian Zhou
- Marine Science & Technology Institute, College of Environmental Science & Engineering, Yangzhou University, 196#, Huayang West Street, Yangzhou, 225127 China
| | - Till F. Schäberle
- Institute for Insect Biotechnology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Branch for Bioresources, 35392 Giessen, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35392 Giessen, Germany
| | - Li Shen
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225001 China
| | - Chang-Yun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237 China
| | - Yang Liu
- Institute for Insect Biotechnology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Branch for Bioresources, 35392 Giessen, Germany
| |
Collapse
|
10
|
Singh K, Gupta JK, Chanchal DK, Shinde MG, Kumar S, Jain D, Almarhoon ZM, Alshahrani AM, Calina D, Sharifi-Rad J, Tripathi A. Natural products as drug leads: exploring their potential in drug discovery and development. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4673-4687. [PMID: 39621089 DOI: 10.1007/s00210-024-03622-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/08/2024] [Indexed: 04/11/2025]
Abstract
Natural products have been pivotal in drug discovery, offering a wealth of bioactive compounds that significantly contribute to therapeutic developments. Despite the rise of synthetic chemistry, natural products continue to play a crucial role due to their unique chemical structures and diverse biological activities. This study reviews and evaluates the potential of natural products in drug discovery and development, emphasizing the integration of traditional knowledge with modern drug discovery methodologies and addressing the associated challenges. A comprehensive literature search was conducted across PubMed/MedLine, Scopus, Web of Science, Google Scholar, and Cochrane Library, covering publications from 2000 to 2023. Inclusion criteria focused on studies related to natural products, bioactive compounds, medicinal plants, phytochemistry, and AI applications in drug discovery. Data were categorized into source, extraction methods, bioactivity assays, and technological advances. The current review underscores the historical and ongoing importance of natural products in drug discovery. Technological advancements in chromatographic and spectroscopic techniques have improved the isolation and structural elucidation of bioactive compounds. AI and machine learning have streamlined the identification and optimization of natural product leads. Challenges such as biodiversity sustainability and development complexities are discussed, alongside innovative approaches like biosynthetic engineering and metagenomics. Natural products remain a vital source of novel therapeutic agents, providing unique chemical diversity and specific biological activities. Integrating traditional knowledge with modern scientific methods is essential for maximizing the potential of natural products in drug discovery. Despite existing challenges, ongoing research and technological advancements are expected to enhance the efficiency and success of natural product-based drug development.
Collapse
Affiliation(s)
- Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Dilip Kumar Chanchal
- Department of Pharmacognosy, College of Pharmacy, SR Group of Institutions, Jhansi, India
| | | | - Shivendra Kumar
- Department of Pharmacology, Rajiv Academy for Pharmacy, Mathura, Uttar Pradesh, India
| | - Divya Jain
- Department of Microbiology, School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India.
| | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, College of Pharmacy, Shaqra University, Dawadimi, Saudi Arabia
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
| | - Anjali Tripathi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| |
Collapse
|
11
|
Du Y, Bian Y, Baecker D, Dhawan G, Semghouli A, Kiss L, Zhang W, Sorochinsky AE, Soloshonok VA, Han J. Fluorine in the Pharmaceutical Industry: FDA-Approved Fluorine-Containing Drugs in 2024. Chemistry 2025; 31:e202500662. [PMID: 40119787 DOI: 10.1002/chem.202500662] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 03/24/2025]
Abstract
Fluorine has become an essential element in the development of modern pharmaceuticals, due to its unique chemical properties that can significantly enhance the biological activity, metabolic stability, and lipophilicity of drug molecules. This review explores recent advancements in the synthesis and application of fluorine-containing drugs approved by the US Food and Drug Administration (FDA) in 2024. These novel drugs demonstrate improved efficacy and safety profiles, addressing a range of therapeutic areas including oncology, infectious diseases, metabolic disorders and genetic disorders that affect the adrenal glands. The incorporation of fluorine atoms into drug candidates has facilitated the development of molecules with optimized pharmacokinetic and pharmacodynamic properties, leading to better patient outcomes. The review further discusses the synthetic methodologies employed, the structural characteristics of these drugs, and their clinical implications, providing insights into the ongoing innovation within the pharmaceutical industry driven by fluorine chemistry.
Collapse
Affiliation(s)
- Youlong Du
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Yeping Bian
- Department of Intensive Care Unit, Geriatric Hospital of Nanjing Medical University, No.30 Luojia Road, Nanjing, 210024, China
| | - Daniel Baecker
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195, Berlin, Germany
| | - Gagan Dhawan
- School of Allied Medical Sciences, Delhi Skill and Entrepreneurship University, Dwarka, New Delhi, 110077, India
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, Kalkaji, New Delhi, 110019, India
| | - Anas Semghouli
- Institute of Organic Chemistry, Stereochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest, H-1117, Hungary
| | - Loránd Kiss
- Institute of Organic Chemistry, Stereochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest, H-1117, Hungary
| | - Wei Zhang
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA, USA, 02125
| | - Alexander E Sorochinsky
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, The National Academy of Sciences of Ukraine, 1 Murmanska str., Kyiv, 02094, Ukraine
| | - Vadim A Soloshonok
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel Lardizábal 3, San Sebastián, 20018, Spain
- IKERBASQUE, Basque Foundation for Science, María Díaz de Haro 3, Plaza Bizkaia, Bilbao, 48013, Spain
| | - Jianlin Han
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| |
Collapse
|
12
|
Xie S, Yang G, Wu J, Jiang L, Yuan C, Xu P, Huang M, Liu Y, Li J. In silico screening of natural products as uPAR inhibitors via multiple structure-based docking and molecular dynamics simulations. J Biomol Struct Dyn 2025; 43:3064-3075. [PMID: 38111151 DOI: 10.1080/07391102.2023.2295386] [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: 10/25/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
Cancer remains one of the most pressing challenges to global healthcare, exerting a significant impact on patient life expectancy. Cancer metastasis is a critical determinant of the lethality and treatment resistance of cancer. The urokinase-type plasminogen activator receptor (uPAR) shows great potential as a target for anticancer and antimetastatic therapies. In this work, we aimed to identify potential uPAR inhibitors by structural dynamics-based virtual screenings against a natural product library on four representative apo-uPAR structural models recently derived from long-timescale molecular dynamics (MD) simulations. Fifteen potential inhibitors (NP1-NP15) were initially identified through molecular docking, consensus scoring, and visual inspection. Subsequently, we employed MD-based molecular mechanics-generalized Born surface area (MM-GBSA) calculations to evaluate their binding affinities to uPAR. Structural dynamics analyses further indicated that all of the top 6 compounds exhibited stable binding to uPAR and interacted with the critical residues in the binding interface between uPAR and its endogenous ligand uPA, suggesting their potential as uPAR inhibitors by interrupting the uPAR-uPA interaction. We finally predicted the ADMET properties of these compounds. The natural products NP5, NP12, and NP14 with better binding affinities to uPAR than the uPAR inhibitors previously discovered by us were proven to be potentially orally active in humans. This work offers potential uPAR inhibitors that may contribute to the development of novel effective anticancer and antimetastatic therapeutics.
Collapse
Affiliation(s)
- Song Xie
- College of Chemistry, Fuzhou University, Fuzhou, China
| | - Guiqian Yang
- College of Chemistry, Fuzhou University, Fuzhou, China
| | - Juhong Wu
- College of Chemistry, Fuzhou University, Fuzhou, China
| | | | - Cai Yuan
- College of Chemistry, Fuzhou University, Fuzhou, China
| | - Peng Xu
- College of Chemistry, Fuzhou University, Fuzhou, China
| | | | - Yichang Liu
- School of Pharmacy, Nantong University, Nantong, China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, China
- Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, Xiamen, China
| |
Collapse
|
13
|
Chen Z, Guo Y, Peng Y, Tan X, Chen H, Luo D, Luo K, Wu D, Huang Z, Yu Z, Tao C. Synthesis and biological evaluation of novel isatin-phenol hybrids as potential antitumor agents. Bioorg Chem 2025; 157:108232. [PMID: 39919326 DOI: 10.1016/j.bioorg.2025.108232] [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: 11/18/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
Chemotherapy was one of indispensable methods for treating cancer, and the development of novel antitumor drugs was necessary due to the emergent drug resistance and undesirable side effects. In the current study, we successfully constructed a novel library of isatin-phenol hybrids by chemical coupling of isatin (1) with a series of active phenols including honokiol (2), magnolol (3), bis(4-hydroxy-3-methylphenyl) sulfide (4), bisphenol A (5), carvacrol (6), and hydroxyqunioline (7) respectively. The target molecules were screened for anticancer activity, and we further investigate the anti-cancer mechanism of the most potent compound IPH10 in vitro and in vivo. Animal experiments demonstrated that IPH10 possessed strong anti-tumor effects in vivo without hepatic and renal toxicity. Moreover, the effects of IPH10 on mitochondrial membrane potential (JC-1) and reactive oxygen species (ROS) in tumor cells were investigated, and the results showed that IPH10 could significantly increase the content of ROS and dramatically decrease the mitochondrial membrane potential in tumor cells. Furthermore, the effect of IPH10 on apoptotic proteins in tumor cells was also explored by Western blotting analysis, which revealed that IPH10 could significantly increase the protein content of cleaved caspase-9/cleaved caspase-3/cleaved caspase-7/cleaved PARP. Taken together, the current study reported a promising novel chemotherapeutic drug candidate IPH10 that could inhibit the growth and induce the apoptosis of tumor cells.
Collapse
Affiliation(s)
- Zhi Chen
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yishan Guo
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yuwei Peng
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Xiaojun Tan
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Haoxiong Chen
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Daqiang Luo
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Kaixuan Luo
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Dudu Wu
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zunnan Huang
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer‑Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| | - Zhiqiang Yu
- Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China.
| | - Cheng Tao
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The First Dongguan Affiliated Hospital and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
14
|
Wang Z, Zhao M, Yu Y, Kong F, Lin N, Wang Q. Marine Fungal Metabolites as Potential Antidiabetic Agents: A Comprehensive Review of Their Structures and Enzyme Inhibitory Activities. Mar Drugs 2025; 23:142. [PMID: 40278263 PMCID: PMC12028496 DOI: 10.3390/md23040142] [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: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Diabetes mellitus has emerged as a global public health crisis, with Type 2 diabetes (T2D) constituting over 90% of cases. Current treatments are palliative, primarily focusing on blood glucose modulation. This review systematically evaluates 181 bioactive compounds isolated from 66 marine fungal strains for their inhibitory activities against key diabetes-related enzymes, including α-glucosidase, protein tyrosine phosphatase 1B (PTP1B), dipeptidyl peptidase-4 (DPP-4), glycogen synthase kinase-3β (GSK-3β), and fatty acid-binding protein 4 (FABP4). These compounds, categorized into polyketides, alkaloids, terpenoids, and lignans, exhibit multitarget engagement and nanomolar-to-micromolar potency. The review highlights the potential of marine fungal metabolites as novel antidiabetic agents, emphasizing their structural novelty and diverse mechanisms of action. Future research should focus on overcoming challenges related to yield and extraction, leveraging advanced technologies such as genetic engineering and synthetic biology to enhance drug development.
Collapse
Affiliation(s)
- Zimin Wang
- Department of Pediatric intensive Care Medicine, Hainan Women and Children’s Medical Center, Haikou 570100, China;
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of of Marine Science and Biotechnology, Guangxi Minzu University, Nanning 530006, China;
| | - Meirong Zhao
- College of Food and Pharmaceutical Engineering, Guangxi Vocational University of Agriculture, Nanning 530006, China;
| | - Yunxia Yu
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of of Marine Science and Biotechnology, Guangxi Minzu University, Nanning 530006, China;
| | - Fandong Kong
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of of Marine Science and Biotechnology, Guangxi Minzu University, Nanning 530006, China;
| | - Nanxin Lin
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of of Marine Science and Biotechnology, Guangxi Minzu University, Nanning 530006, China;
| | - Qi Wang
- Department of Pediatric intensive Care Medicine, Hainan Women and Children’s Medical Center, Haikou 570100, China;
| |
Collapse
|
15
|
Chen K, Wang S, Fu S, Kim J, Park P, Liu R, Lei K. 4(3 H)-Quinazolinone: A Natural Scaffold for Drug and Agrochemical Discovery. Int J Mol Sci 2025; 26:2473. [PMID: 40141117 PMCID: PMC11941892 DOI: 10.3390/ijms26062473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
4(3H)-quinazolinone is a functional scaffold that exists widely both in natural products and synthetic organic compounds. Its drug-like derivatives have been extensively synthesized with interesting biological features including anticancer, anti-inflammatory, antiviral, antimalarial, antibacterial, antifungal, and herbicidal, etc. In this review, we highlight the medicinal and agrochemical versatility of the 4(3H)-quinazolinone scaffold according to the studies published in the past six years (2019-2024), and comprehensively give a summary of the target recognition, structure-activity relationship, and mechanism of its analogs. The present review is expected to provide valuable guidance for discovering novel lead compounds containing 4(3H)-quinazolinone moiety in both drug and agrochemical research.
Collapse
Affiliation(s)
- Ke Chen
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Shumin Wang
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.W.); (S.F.)
| | - Shuyue Fu
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.W.); (S.F.)
| | - Junehyun Kim
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Phumbum Park
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Rui Liu
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Kang Lei
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.W.); (S.F.)
| |
Collapse
|
16
|
Urbanska N, Ashaolu TJ, Mattova S, Simko P, Kiskova T. The Potential of Selected Plants and Their Biologically Active Molecules in the Treatment of Depression and Anxiety Disorders. Int J Mol Sci 2025; 26:2368. [PMID: 40076986 PMCID: PMC11900588 DOI: 10.3390/ijms26052368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
The incidence of anxiety and depression disorders is increasing worldwide. There is an increasing incidence of hard-to-treat depression with various aspects of origin. Almost 80% of people prefer to use natural remedies and supplements as their primary healthcare solution. Not surprisingly, around one-third of drugs were inspired by nature. Over the past three decades, the use of such remedies has increased significantly. Synthetic antidepressants may cause various negative side effects, whereas herbal medicines are favored because of their ability to relieve symptoms with minimal to no side effects and lower financial burden. This review provides an overview of herbs and biologically active compounds used to treat depression.
Collapse
Affiliation(s)
- Nicol Urbanska
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Safarik University in Kosice, Srobarova 2, 041 54 Kosice, Slovakia
| | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, 254 Nguyen Van Linh Street, Thanh-Khe District, Da Nang 550000, Vietnam
| | - Simona Mattova
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Safarik University in Kosice, Srobarova 2, 041 54 Kosice, Slovakia
| | - Patrik Simko
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Safarik University in Kosice, Srobarova 2, 041 54 Kosice, Slovakia
| | - Terezia Kiskova
- Institute of Pathology, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, Rastislavova 43, 040 01 Kosice, Slovakia
| |
Collapse
|
17
|
Ali S, Shaikh S, Ahmad K, Choi I. Identification of active compounds as novel dipeptidyl peptidase-4 inhibitors through machine learning and structure-based molecular docking simulations. J Biomol Struct Dyn 2025; 43:1611-1620. [PMID: 38100571 DOI: 10.1080/07391102.2023.2292299] [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: 07/25/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
The enzyme dipeptidyl peptidase 4 (DPP4) is a potential therapeutic target for type 2 diabetes (T2DM). Many synthetic anti-DPP4 medications are available to treat T2DM. The need for secure and efficient medicines has been unmet due to the adverse side effects of existing DPP4 medications. The present study implemented a combined approach to machine learning and structure-based virtual screening to identify DPP4 inhibitors. Two ML models were trained based on DPP4 IC50 datasets. The ML models random forest (RF) and multilayer perceptron (MLP) neural network showed good accuracy, with the area under the curve being 0.93 and 0.91, respectively. The natural compound library was screened through ML models, and 1% (217) of compounds were selected for further screening. Structure-based virtual screening was performed along with positive control sitagliptin to obtain more specific and selective leads for DPP4. Based on binding affinity, drug-likeness properties, and interaction with DPP4, Z-614 and Z-997 compounds showed high binding affinity and specificity in the catalytic pocket of DPP4. Finally, the stability conformation of the DPP4 enzyme complex was checked by a molecular dynamics (MD) simulation. The MD simulation showed that both compounds bind better in the catalytic pocket, but the Z-614 compound altered the DPP4 native conformation. Therefore, Z-614 showed a high deviation in the backbone. This combined approach (ML and structure-based) study reported that Z-997 binds most stably to DPP4 in their catalytic pocket with a binding free energy of -70.3 kJ/mol, suggesting its therapeutic potential as a treatment option for T2DM disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
18
|
Cai S, Guo X, Yang H, Zhao T, Li Y, Deng N, Gao Z, Meng Q, Li X, Wang S. Synthesis and antitumor effects of novel betulinic acid derivatives bearing electrophilic moieties. Bioorg Med Chem 2025; 119:118062. [PMID: 39756343 DOI: 10.1016/j.bmc.2025.118062] [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: 10/10/2024] [Revised: 12/21/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
Betulinic acid (BA) is a kind of naturally occurring lupane pentacyclic triterpenoid, possessing various biological activities including antiviral, anti-inflammatory and antitumor activity. Covalent inhibitors, characterized by electrophilic warheads that form covalent bonds with specific amino acid residues of target proteins, have garnered enormous attention in anticancer agent discovery over the past decade owing to their exceptional selectivity and efficacy. In this study, BA was structurally modified with electrophilic groups, and 23 derivatives of BA were synthesized. Most of these BA derivatives exhibited improved antiproliferative activity against MCF-7, HeLa, MDA-MB-231 cells in MTT assay, especially the compound 15b (IC50 = 1.09 μM against MCF-7 cells). Further study demonstrated that 15b inhibited the migration and clone formation of MCF-7 cells, induced the apoptosis, autophagy and cycle arrest at G2/M phase in MCF-7 cells, and promoted the production of intracellular reactive oxygen species (ROS). Western blot analysis showed that 15b inhibited AKT/mTOR signaling pathway in MCF-7 cells. In addition, 15b reversed the resistance of JIMT-1 cells to trastuzumab, which might be related to the inhibition of AKT/mTOR pathway. Finally, 15b significantly inhibited the growth of tumor in the breast cancer xenograft mouse model with 36 % inhibition rate of tumor growth and without significant reduction of mouse body weight.
Collapse
Affiliation(s)
- Sen Cai
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xiuhan Guo
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Haozhe Yang
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tianyu Zhao
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yueqing Li
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Ning Deng
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Zhigang Gao
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Qingwei Meng
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Xiaorui Li
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| | - Shisheng Wang
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China.
| |
Collapse
|
19
|
Luo YY, Xu JB, Huang S, Zhou XL. Diterpenoid alkaloids from the roots of Aconitum bulbilliferum. Fitoterapia 2025; 181:106387. [PMID: 39788436 DOI: 10.1016/j.fitote.2025.106387] [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: 10/17/2024] [Revised: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The phytochemistry study of Aconitum bulbilliferum Hand.-Mazz. is firstly reported. Two C20-diterpenoid alkaloids, named bulbilliferines A and B (1 and 2), three C19-diterpenoid alkaloids, bulbillifedines A-C (3-5), and twenty known compounds (6-25) were isolated from the roots of A. bulbilliferum. The structures of these compounds were determined by the comprehensive spectroscopic analyses (HRESIMS, IR, 1D and 2D NMR). Among them, bulbilliferine A (1) is a denudatine-type diterpenoid alkaloid bearing a 16, 17-epoxy group. At a nontoxic dose of 10 mg/kg, 14-O-anisoylchasmanine (14) hydrochloride and austroconitine B (17) hydrochloride respectively exhibited the excellent analgesic effects with 83.6 % and 83.1 % inhibitions against acetic acid-induced writhing of mice.
Collapse
Affiliation(s)
- Yan-Yan Luo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, PR China; Key laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, Sichuan, PR China
| | - Jin-Bu Xu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, PR China
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, PR China
| | - Xian-Li Zhou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, PR China; Key laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, Sichuan, PR China.
| |
Collapse
|
20
|
Jiang Y, Tang Z, Jiang M, Wang J, Wang Y. Small Molecule Inhibitors Targeting Cdc2-Like Kinase 4: Advances, Challenges, and Opportunities. Chem Biol Drug Des 2025; 105:e70087. [PMID: 40095342 DOI: 10.1111/cbdd.70087] [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: 10/20/2024] [Revised: 02/03/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025]
Abstract
Cdc2-like kinase 4 (Clk4), a key member of the CMGC kinase family, plays a crucial role in alternative splicing, which profoundly influences various physiological processes, including cellular signaling, proliferation, and survival. Its involvement in these vital functions has positioned Clk4 as an important target for therapeutic intervention in a range of diseases, such as neurodegenerative disorders, viral and parasitic infections, and cancer. This review highlights recent advancements in Clk4 inhibitors, covering both natural, and synthetic compounds. It further examines the core scaffolds and essential functional groups of Clk4 small-molecule inhibitors, emphasizing the most promising chemical structures. Additionally, the review explores the structure-activity relationships (SARs) and molecular binding modes of existing Clk4 inhibitors, offering insights and strategies for the development of novel Clk4-targeted drugs. This review highlights recent advancements in small molecule inhibitors targeting Clk4, emphasizing their potential in treating cancers and neurodegenerative diseases. It explores SARs, binding modes, and challenges in developing selective Clk4 inhibitors, offering insights for future therapeutic strategies.
Collapse
Affiliation(s)
- Yu Jiang
- West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zihua Tang
- West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Minggao Jiang
- West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jing Wang
- Pediatric Otolaryngology Head and Neck Surgery, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanhai Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
21
|
Vinh LB, Lee KS, Han YK, Kim YJ, Kim S, Shah AB, Byun Y, Lee KY. Allergy Inhibition Using Naturally Occurring Compounds Targeting Thymic Stromal Lymphopoietin Pathways: a Comprehensive Review. Biomol Ther (Seoul) 2025; 33:249-267. [PMID: 39933953 PMCID: PMC11893497 DOI: 10.4062/biomolther.2024.177] [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: 09/24/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 02/13/2025] Open
Abstract
Naturally occurring compounds have widely been applied to treat diverse pharmacological effects, including asthma, allergic diseases, antioxidants, inflammation, antibiotics, and cancer. Recent research has revealed the essential role of the thymic stromal lymphopoietin (TSLP) in regulating inflammatory responses at mucosal barriers and maintaining immune homeostasis. Asthma, inflammation, and chronic obstructive pulmonary disease are allergic disorders in which TSLP plays a significant role. Although TSLP's role in type 2 immune responses has undergone comprehensive investigation, its involvement in inflammatory diseases and cancer has also been found to be expanding. However, investigating how to block the TSLP pathway using natural products has been limited. This paper summarizes the roles of various medicinal plants and their chemical components that effectively inhibit the TSLP pathway. In addition, we also highlight the contributions of several plant-derived compounds to treat allergic diseases via targeting TSLP. This review intends to offer innovative concepts to scientists investigating the use of naturally produced compounds and extracts for the treatment of allergic illnesses.
Collapse
Affiliation(s)
- Le Ba Vinh
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, Hanoi 11355, Vietnam
| | - Kyeong Seon Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Yoo Kyong Han
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Young Jun Kim
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Suzy Kim
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Abdul Bari Shah
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Ki Yong Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
22
|
Gangwal A, Lavecchia A. Artificial Intelligence in Natural Product Drug Discovery: Current Applications and Future Perspectives. J Med Chem 2025; 68:3948-3969. [PMID: 39916476 PMCID: PMC11874025 DOI: 10.1021/acs.jmedchem.4c01257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 12/01/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Drug discovery, a multifaceted process from compound identification to regulatory approval, historically plagued by inefficiencies and time lags due to limited data utilization, now faces urgent demands for accelerated lead compound identification. Innovations in biological data and computational chemistry have spurred a shift from trial-and-error methods to holistic approaches to medicinal chemistry. Computational techniques, particularly artificial intelligence (AI), notably machine learning (ML) and deep learning (DL), have revolutionized drug development, enhancing data analysis and predictive modeling. Natural products (NPs) have long served as rich sources of biologically active compounds, with many successful drugs originating from them. Advances in information science expanded NP-related databases, enabling deeper exploration with AI. Integrating AI into NP drug discovery promises accelerated discoveries, leveraging AI's analytical prowess, including generative AI for data synthesis. This perspective illuminates AI's current landscape in NP drug discovery, addressing strengths, limitations, and future trajectories to advance this vital research domain.
Collapse
Affiliation(s)
- Amit Gangwal
- Department
of Natural Product Chemistry, Shri Vile
Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, 424001 Maharashtra, India
| | - Antonio Lavecchia
- “Drug
Discovery” Laboratory, Department of Pharmacy, University of Naples Federico II, I-80131 Naples, Italy
| |
Collapse
|
23
|
Ma X, Yang Z, Luo Y, Jin Z, Zou J, Wang Y, Zhao X. A novel fluorescent probe with Aggregation-Induced emission characteristics for PTP1B activity sensing and inhibitor screening. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125394. [PMID: 39520822 DOI: 10.1016/j.saa.2024.125394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/21/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is an attractive target for the treatment of metabolic diseases such as type 2 diabetes and obesity. In this study, a novel fluorescent probe with aggregation-induced emission (AIE) characteristics was designed and synthesized. Within the fluorescent probe, a tetraphenylethene core is connected to a peptide sequence that can be specifically recognized and hydrolysed by PTP1B. Due to the dephosphorylation of PTP1B, the fluorescent probe exhibited AIE in a turn-on manner, indicating PTP1B activity. This probe was successfully used to detect PTP1B activity in HepG2 cell lysates. Then, a probe-based method was applied to screen for potential PTP1B inhibitors from a natural product library, and three novel PTP1B inhibitors were discovered. These findings indicated that the proposed approach offered a new avenue for discovering potential PTP1B inhibitors.
Collapse
Affiliation(s)
- Xiangwei Ma
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhenzhong Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Chinese Medicine Modernization, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanlin Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zehua Jin
- State Key Laboratory of Chinese Medicine Modernization, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jingtao Zou
- Tonghua Huaxia Pharmaceutical Company, Tonghua 134000, China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Chinese Medicine Modernization, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Xiaoping Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
24
|
Jung E, Griesser T, Costafrolaz J, Duverger O, Mattenberger Y, Dittmann S, Dorst A, Major A, Dailler D, Schäfle D, Sievers S, Brodolin K, Viollier PH, Sander P, Gademann K. Switching Residues: A Platform for the Synthesis of Fidaxomicin Antibiotics. Angew Chem Int Ed Engl 2025; 64:e202419095. [PMID: 39584779 PMCID: PMC11811690 DOI: 10.1002/anie.202419095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Peripheral modification is often the main approach to optimize natural products for improved biological activity or desired physicochemical properties. This procedure inevitably increases molecular weight, often accompanied by undesired increased lipophilicity. Removing structural elements from natural products is not always tolerated. This is also the case for the antibiotic fidaxomicin (Fdx), where every structural component has been shown to be crucial for antibiotic activity. In this work, we demonstrate how the residue switching approach can maintain biological activity of Fdx derivatives by replacing the rhamnoside-dichlorohomoorsellinate moiety of Fdx with smaller, more polar building blocks. We used palladium-catalysed allylic substitution to selectively install N-nucleophiles on the core of Fdx. The new derivatives were designed to mimic the binding of Fdx to the bacterial RNA polymerase. Evaluation against Mycobacterium tuberculosis, Clostridioides difficile, and the Gram-negative model organism Caulobacter crescentus demonstrated that the newly introduced residues can restore antibiotic activity, which was further supported by on-target RNA polymerase assays. We combined the allylic substitution with an organocatalysed novioside acylation protocol to enable the functionalisation of two vectors on Fdx in one pot. This platform greatly expands the accessible chemical space for Fdx derivatives and enables the future development of systemic Fdx antibiotics.
Collapse
Affiliation(s)
- Erik Jung
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - Tizian Griesser
- Institute of Medical MicrobiologyUniversity of ZurichZurichSwitzerland
| | - Jordan Costafrolaz
- Department of Microbiology and Molecular MedicineFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Ondine Duverger
- Institut de Recherche en Infectiologie de MontpellierUniv. MontpellierCNRSMontpellier34293France
| | - Yves Mattenberger
- Department of Microbiology and Molecular MedicineFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Silvia Dittmann
- Department of Microbial Physiology and Molecular BiologyInstitute of MicrobiologyCenter for Functional Genomics of MicrobesUniversity of GreifswaldGreifswaldGermany
| | - Andrea Dorst
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - Alexander Major
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - David Dailler
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - Daniel Schäfle
- Institute of Medical MicrobiologyUniversity of ZurichZurichSwitzerland
| | - Susanne Sievers
- Department of Microbial Physiology and Molecular BiologyInstitute of MicrobiologyCenter for Functional Genomics of MicrobesUniversity of GreifswaldGreifswaldGermany
| | - Konstantin Brodolin
- Institut de Recherche en Infectiologie de MontpellierUniv. MontpellierCNRSMontpellier34293France
| | - Patrick H. Viollier
- Department of Microbiology and Molecular MedicineFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Peter Sander
- Institute of Medical MicrobiologyUniversity of ZurichZurichSwitzerlandNational Reference Laboratory for MycobacteriaUniversity of ZurichZurichSwitzerland
| | - Karl Gademann
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| |
Collapse
|
25
|
Rana A, Mishra A, Awasthi SK. Recent advancements in the chemistry of Diels-Alder reaction for total synthesis of natural products: a comprehensive review (2020-2023). RSC Adv 2025; 15:4496-4525. [PMID: 39931410 PMCID: PMC11808662 DOI: 10.1039/d4ra07989b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Despite being discovered nearly a century ago, the Diels-Alder (DA) reaction remains a crucial tool in the total synthesis of natural products. It accommodates a broad range of building blocks with varying complexity and levels of derivatization, allowing the formation of six-membered rings with precise stereochemistry. This, in turn, simplifies the synthesis of core structures found in many natural products. In recent years, modifications to the traditional Diels-Alder reaction have expanded its scope. These modifications include the inverse electron demand Diels-Alder reaction, dehydro Diels-Alder reaction, hetero-Diels-Alder reaction, photoenolization Diels-Alder reaction, asymmetric Diels-Alder reaction, and domino Diels-Alder reaction have been employed to extend the scope of this process in the synthesis of natural products. This review discusses the application of the Diels-Alder reaction in the total synthesis of natural products from 2020 to 2023, along with select methodologies that are inspired by or can be used to synthesize natural products.
Collapse
Affiliation(s)
- Anitesh Rana
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi Delhi 110007 India
| | - Anupam Mishra
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi Delhi 110007 India
| | - Satish K Awasthi
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi Delhi 110007 India
| |
Collapse
|
26
|
Lentschat H, Liessmann F, Tydings C, Schermeng T, Stichel J, Urban N, Schaefer M, Meiler J, Beck-Sickinger AG. Hederagenin is a Highly Selective Antagonist of the Neuropeptide FF Receptor 1 that Reveals Mechanisms for Subtype Selectivity. Angew Chem Int Ed Engl 2025; 64:e202417786. [PMID: 39641914 DOI: 10.1002/anie.202417786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
RF-amide peptide receptors including the neuropeptide FF receptor 1 (NPFFR1) are G protein-coupled receptors (GPCRs) that modulate diverse physiological functions. High conservation of endogenous ligands and receptors makes the identification of selective ligands challenging. Previously identified antagonists mimic the C-terminus of peptide ligands and lack selectivity towards the closely related neuropeptide FF receptor 2 (NPFFR2) or the neuropeptide Y1 receptor (Y1R). In a high-throughput screening, we identified the pentacyclic triterpenoid hederagenin (1) as a novel selective antagonist for the NPFFR1. Hederagenin (1) is a natural product isolated from Hedera helix (ivy). We characterized its mode of activity using in vitro and in silico methods, revealing an overlapping binding site of the small molecule with the orthosteric peptide agonists. Despite the high similarity of the orthosteric binding pockets of NPFFR1 and NPFFR2, hederagenin (1) shows strong subtype selectivity, particularly caused by slight differences in the shape of the binding pockets and the rigidity of the small molecule. Several residues inhibiting the activity of hederagenin (1) at the NPFFR2 were identified. As NPFFR1 antagonists are discussed as potential candidates for the treatment of chronic pain, these insights into the structural determinants governing subtype specificity will facilitate the development of next-generation analgesics with improved safety and efficacy.
Collapse
Affiliation(s)
- Hannah Lentschat
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| | - Fabian Liessmann
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, 04103, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, ScaDS.AI Dresden/Leipzig, Leipzig, 04105, Germany
| | - Claiborne Tydings
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, 37235, United States
| | - Tina Schermeng
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| | - Jan Stichel
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| | - Nicole Urban
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University, Leipzig, 04107, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University, Leipzig, 04107, Germany
| | - Jens Meiler
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, 04103, Germany
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, 37235, United States
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, 37232, United States
- Center for Scalable Data Analytics and Artificial Intelligence, ScaDS.AI Dresden/Leipzig, Leipzig, 04105, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| |
Collapse
|
27
|
Zhao C, Qin J, Zhang D, Li X, Yang N, Gao T, Song J, Song Y, Huang S, Xu H. NGR-poly(2-ethyl-2-oxazoline)-cholesteryl methyl carbonate enhances the antitumor effect of quercetin liposomes in triple-negative breast cancer. Pharm Dev Technol 2025; 30:137-149. [PMID: 39764693 DOI: 10.1080/10837450.2025.2450434] [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: 10/14/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
In this paper, the pH-sensitive targeting functional material NGR-poly(2-ethyl-2-oxazoline)-cholesteryl methyl carbonate (NGR-PEtOz-CHMC, NPC) modified quercetin (QUE) liposomes (NPC-QUE-L) was constructed. The structure of NPC was confirmed by infrared spectroscopy (IR) and nuclear magnetic resonance hydrogen spectrum (1H-NMR). Pharmacokinetic results showed that the accumulation of QUE in plasma of the NPC-QUE-L group was 1.28 times and 2.43 times that of the QUE Solution and QUE-L groups, respectively. The release amount of NPC-QUE-L in an acidic environment was significantly higher than in physiological pH value. The order of the tumor cell inhibition rate in different pH environments was NPC-QUE-L > PC-QUE-L > QUE-L. In addition, the cellular uptake of NPC-modified liposomes was higher than that of PC-modified and unmodified liposomes, indicating that NPC had good pH-sensitivity and targeting. In the triple-negative breast cancer (TNBC) model, the relative tumor proliferation rate of NPC-QUE-L is about 73%, which is better than that of the QUE solution group. Western blot results show that NPC-QUE-L can effectively reduce the expression of α-smooth actin and transforming growth factor-β1 in tumor tissues, and improve the degree of tumor fibrosis. In this study, NPC could endow QUE liposomes with good stability, pH-sensitivity, and targeting, which provides a reference for improving the solubility and targeting of poorly soluble natural drug components.
Collapse
Affiliation(s)
- Chengcheng Zhao
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Jian Qin
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Dingyu Zhang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Xue Li
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Ning Yang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Tingyu Gao
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Junliang Song
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Yule Song
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Shouzhen Huang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| | - Huan Xu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, China
| |
Collapse
|
28
|
Rose TM, Baranek M, Kaka M, Shwani S. Natural drugs: Trends, properties, and decline in FDA approvals. J Pharm Sci 2025; 114:782-786. [PMID: 39730033 DOI: 10.1016/j.xphs.2024.12.007] [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: 08/13/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024]
Abstract
The purpose of this review is to better characterize the contribution and properties of FDA-approved drugs that can be found unmodified in nature. Defined inclusion criteria were applied to drugs identified in previous studies and in annual FDA approval reports to compile a comprehensive list of approved drugs found in nature. Databases and scientific literature were searched to identify chemical and drug properties of these entities, including chemical classes, approval years, drug indications, and approved delivery methods. A random sample of FDA-approved drugs not found in nature was also created for comparison. Drugs from nature are estimated to represent 5 % of FDA approved drugs. The most common classes of natural product drugs are alkaloids, oligopeptides, polyphenols, and polyketides. Approvals of unmodified drugs from nature have declined at a rate of about two per decade since the mid-1900s. Compared to non-natural drugs, drugs from nature are more likely to be used as antibacterials and for dermatological conditions. Natural drugs are also less likely to be delivered orally and more likely to have narrow therapeutic indexes. Given the limitations of unmodified natural products as drugs, the pharmaceutical sciences will likely continue to play an important role in improving the drug-likeness of natural scaffolds.
Collapse
Affiliation(s)
- Tyler M Rose
- Roseman University of Health Sciences, College of Pharmacy, 10920 South River Front Parkway, South Jordan, UT 84095, USA.
| | - Monika Baranek
- Roseman University of Health Sciences, College of Pharmacy, 10920 South River Front Parkway, South Jordan, UT 84095, USA.
| | - Maryam Kaka
- Roseman University of Health Sciences, College of Pharmacy, 10920 South River Front Parkway, South Jordan, UT 84095, USA.
| | - Sarah Shwani
- Roseman University of Health Sciences, College of Pharmacy, 10920 South River Front Parkway, South Jordan, UT 84095, USA.
| |
Collapse
|
29
|
Baz MM, El-Tabakh MAM, Selim A, Alasmari SM, Alkhaibari AM, Alruhaili MH, Gattan HS, Abdelkhalek HF. Chemical composition and bio-efficacy of agro-waste plant extracts and their potential as bioinsecticides against Culex pipiens mosquitoes. Parasitol Int 2025; 104:102968. [PMID: 39271003 DOI: 10.1016/j.parint.2024.102968] [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: 08/16/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Mosquitoes are considered one of the most lethal creatures on the planet and are responsible for millions of fatalities annually through the transmission of several diseases to humans. Green trash is commonly employed in agricultural fertilizer manufacturing and microbial bioprocesses for energy production. However, there is limited information available on the conversion of green waste into biocides. This study investigates the viability of utilizing green waste as a new biopesticide against Culex pipiens mosquito larvae. The current study found that plant extracts from Punica granatum (98.4 % mortality), Citrus sinensis (92 % mortality), Brassica oleracea (88 % mortality), Oryza sativa (81.6 % mortality), and Colocasia esculenta (53.6 % mortality) were very good at killing Cx. pipiens larvae 24 h post-treatment. The LC50 values were 314.43, 370.72, 465.59, 666.67, and 1798.03 ppm for P. granatum, C. sinensis, B. oleracea, O. sativa, and C. esculenta, respectively. All plant extracts, particularly P. granatum extract (14.93 and 41.87 U/g), showed a significant reduction in acid and alkaline phosphate activity. Additionally, pomegranate extract showed a significant decrease (90 %) in field larval density, with a stability of up to five days post-treatment. GC-MS results showed more chemical classes, such as terpenes, esters, fatty acids, alkanes, and phenolic compounds. HPLC analysis revealed that the analyzed extracts had a high concentration of phenolic and flavonoid components. Moreover, there are many variations among these plants in the amount of each compound. The docking interaction showed a simulation of the atomic-level interaction between a protein and a small molecule through the binding site of target proteins, explaining the most critical elements influencing the enzyme's activity or inhibitions. The study's findings showed that the various phytochemicals found in agro-waste plants had high larvicidal activity and provide a safe and efficient substitute to conventional pesticides for pest management, as well as a potential future in biotechnology.
Collapse
Affiliation(s)
- Mohamed M Baz
- Entomology Department, Faculty of Science, Benha University, Benha 13518, Egypt.
| | | | - Abdelfattah Selim
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Saeed M Alasmari
- Department of Biology, Faculty of Science and Arts, Najran University, 1988 Najran, Saudi Arabia
| | - Abeer Mousa Alkhaibari
- Department of Biology, Faculty of Science, University of Tabuk, 71491 Tabuk, Saudi Arabia
| | - Mohammed H Alruhaili
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Special Infectious Agents Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Hattan S Gattan
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; Special Infectious Agents Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Heba F Abdelkhalek
- Entomology Department, Faculty of Science, Benha University, Benha 13518, Egypt
| |
Collapse
|
30
|
Wu XY, Dong QW, Zhang YB, Li JX, Zhang MQ, Zhang DQ, Cui YL. Cimicifuga heracleifolia kom. Attenuates ulcerative colitis through the PI3K/AKT/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118892. [PMID: 39395768 DOI: 10.1016/j.jep.2024.118892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cimicifuga heracleifolia Kom. (C. heracleifolia) has demonstrated efficacy in treating gastrointestinal disorders, including splenasthenic diarrhea. Ulcerative colitis (UC), a chronic inflammatory bowel disease, shares similarities with splenasthenic diarrhea. However, the pharmacological effects of C. heracleifolia on UC and the underlying mechanisms remain unexplored. AIM OF THE STUDY The present study investigates the therapeutic potential and mechanisms of C. heracleifolia in UC. METHODS Initially, network pharmacology analysis, encompassing ingredient screening, target prediction, protein-protein interaction (PPI) network analysis, and enrichment analysis, was employed to predict the mechanisms of C. heracleifolia. The findings were further validated using transcriptomics and functional assays in a dextran sulfate sodium (DSS)-induced UC model. Additionally, bioactive compounds were identified through surface plasmon resonance (SPR) analysis, molecular docking, and cell-based assays. RESULTS A total of 52 ingredients of C. heracleifolia were screened, and 32 key targets were identified within a PPI network comprising 285 potential therapeutic targets. Enrichment analysis indicated that the anti-UC effects of C. heracleifolia are mediated through immune response modulation and the inhibition of inflammatory signaling pathways. In vivo experiments showed that C. heracleifolia mitigated histological damage in the colon, reduced the expression of phosphorylated Akt1, nuclear factor-kappa B (NF-κB) p65, and inhibitor of Kappa B kinase α/β (IKKα/β), suppressed the content of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), and enhanced the expression of tight junction proteins. Moreover, cimigenoside, caffeic acid, and methyl caffeate were identified as the bioactive constituents responsible for the UC treatment effects of C. heracleifolia. CONCLUSIONS In summary, this study is the first to demonstrate that C. heracleifolia exerts therapeutic effects on UC by enhancing the intestinal mucosal barrier and inhibiting the phosphatidylinositol 3-kinase (PI3K)/AKT/NF-κB signaling pathway. These findings offer valuable insights into the clinical application of C. heracleifolia for UC management.
Collapse
Affiliation(s)
- Xue-Yi Wu
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yong-Bo Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Jia-Xin Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Mei-Qing Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - De-Qin Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
31
|
Stolarczyk EU, Strzempek W, Muszyńska M, Kubiszewski M, Witkowska AB, Trzcińska K, Wojdasiewicz P, Stolarczyk K. Preparation of Diosgenin-Functionalized Gold Nanoparticles: From Synthesis to Antitumor Activities. Int J Mol Sci 2025; 26:1088. [PMID: 39940856 PMCID: PMC11817374 DOI: 10.3390/ijms26031088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer ranks among the top causes of illness and death globally. Nanotechnology holds considerable promise for enhancing the effectiveness of therapeutic and diagnostic approaches in cancer treatment. Our study presents a promising strategy for applying thiocompound nanomedicine in cancer therapy. Our first study aimed to investigate the biological properties of a new compound thiodiosgenin (TDG)-a new derivative of diosgenin-a natural compound with known antioxidant and anticancer properties. Our current second study aimed to compare the therapeutic efficacy of a new diosgenin-functionalized gold nanoparticles-with its precursor on prostate cancer (DU-145) cell lines. Moreover, the safety of the new thio-derivative and new conjugates was tested against the human epithelial line PNT-2. New advanced analytical techniques were developed for the characterization of nanomaterials using methods such as SP-ICP-MS, UV-Vis, TEM, NMR, FT-IR ELS, and TGA. Our synthetic approach was based, on the one hand, on the ligand exchange of citrates to thiodiosgenin (TDG) on gold nanoparticles, and on the other hand, on the attachment of DG through an ester bond to the linker, which was 3-mercaptopropionic acid (MPA) on gold nanoparticles. Initial in vitro studies indicate that TDG shows greater cytotoxic effects on cancer cells but poses risks to normal prostate epithelial cells (PNT-2). It was demonstrated that all the conjugates produced exhibited significant cytotoxic effects against cancer cells while being less harmful to normal prostate epithelial cells (PNT-2) compared to TDG itself. All the obtained conjugates showed antitumor properties; however, for targeted transport, the system referred to as AuNPs-MPAm1-DG is promising, due to the size of the nanoparticles of 53 nm, zeta potential of -30 mV, and loading content of 27.6%. New methods for synthesizing conjugates with diosgenin were developed and optimized for medical applications. Advanced new analytical methodologies were developed to characterize new conjugates, particularly the use of SP-ICP-MS, to solve existing differences in the shape and morphology of the surface of new conjugates.
Collapse
Affiliation(s)
- Elżbieta U. Stolarczyk
- Spectrometric Methods Department, National Medicine Institute, 30/34 Chełmska Street, 00-725 Warsaw, Poland; (E.U.S.); (A.B.W.)
| | - Weronika Strzempek
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland;
| | - Magdalena Muszyńska
- Faculty of Chemistry, University of Warsaw, 1 Pasteura Street, 02-093 Warsaw, Poland;
| | - Marek Kubiszewski
- Analytical Research Section, Pharmaceutical Analysis Laboratory Łukasiewicz Research Network, Industrial Chemistry Institute, 8 Rydygiera Street, 01-793 Warsaw, Poland; (M.K.); (K.T.)
| | - Anna B. Witkowska
- Spectrometric Methods Department, National Medicine Institute, 30/34 Chełmska Street, 00-725 Warsaw, Poland; (E.U.S.); (A.B.W.)
| | - Kinga Trzcińska
- Analytical Research Section, Pharmaceutical Analysis Laboratory Łukasiewicz Research Network, Industrial Chemistry Institute, 8 Rydygiera Street, 01-793 Warsaw, Poland; (M.K.); (K.T.)
| | - Piotr Wojdasiewicz
- Department of Biophysics, Physiology and Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland;
| | - Krzysztof Stolarczyk
- Faculty of Chemistry, University of Warsaw, 1 Pasteura Street, 02-093 Warsaw, Poland;
| |
Collapse
|
32
|
Nelson S, Parkinson EI. Synthetic-bioinformatic natural product-inspired peptides. Nat Prod Rep 2025; 42:50-66. [PMID: 39479929 PMCID: PMC11525955 DOI: 10.1039/d4np00043a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Indexed: 11/02/2024]
Abstract
Covering: 2016 to 2024Natural products, particularly cyclic peptides, are a promising source of bioactive compounds. Nonribosomal peptide synthetases (NRPSs) play a key role in biosynthesizing these compounds, which include antibiotic and anticancer agents, immunosuppressants, and others. Traditional methods of discovering natural products have limitations including cryptic biosynthetic gene clusters (BGCs), low titers, and currently unculturable organisms. This has prompted the exploration of alternative approaches. Synthetic-bioinformatic natural products (syn-BNPs) are one such alternative that utilizes bioinformatics techniques to predict nonribosomal peptides (NRPs) followed by chemical synthesis of the predicted peptides. This approach has shown promise, resulting in the discovery of a variety of bioactive compounds including peptides with antibacterial, antifungal, anticancer, and proteasome-stimulating activities. Despite the success of this approach, challenges remain especially in the accurate prediction of fatty acid incorporation, tailoring enzyme modifications, and peptide release mechanisms. Further work in these areas will enable the discovery of many bioactive peptides that are currently inaccessible.
Collapse
Affiliation(s)
- Samantha Nelson
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Elizabeth I Parkinson
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47906, USA.
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA
| |
Collapse
|
33
|
Zamani M, Melnychuk T, Eisenhauer A, Gäbler R, Schultz C. Investigating Past, Present, and Future Trends on Interface Between Marine and Medical Research and Development: A Bibliometric Review. Mar Drugs 2025; 23:34. [PMID: 39852536 PMCID: PMC11766621 DOI: 10.3390/md23010034] [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: 12/12/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
The convergence of marine sciences and medical studies has the potential for substantial advances in healthcare. This study uses bibliometric and topic modeling studies to map the progression of research themes from 2000 to 2023, with an emphasis on the interdisciplinary subject of marine and medical sciences. Building on the global publication output at the interface between marine and medical sciences and using the Hierarchical Dirichlet Process, we discovered dominating research topics during three periods, emphasizing shifts in research focus and development trends. Our data show a significant rise in publication output, indicating a growing interest in using marine bioresources for medical applications. The paper identifies two main areas of active research, "natural product biochemistry" and "trace substance and genetics", both with great therapeutic potential. We used social network analysis to map the collaborative networks and identify the prominent scholars and institutions driving this research and development progress. Our study indicates important paths for research policy and R&D management operating at the crossroads of healthcare innovation and marine sciences. It also underscores the significance of quantitative foresight methods and interdisciplinary teams in identifying and interpreting future scientific convergences and breakthroughs.
Collapse
Affiliation(s)
- Mehdi Zamani
- Kiel Institute for Responsible Innovation, Chair of Technology Management, Kiel University (Christian-Albrechts-Universität zu Kiel), 24118 Kiel, Germany; (M.Z.); (T.M.)
| | - Tetyana Melnychuk
- Kiel Institute for Responsible Innovation, Chair of Technology Management, Kiel University (Christian-Albrechts-Universität zu Kiel), 24118 Kiel, Germany; (M.Z.); (T.M.)
| | - Anton Eisenhauer
- GEOMAR Helmholtz-Zentrum für Ozeanforschung Kiel, 24148 Kiel, Germany; (A.E.); (R.G.)
| | - Ralph Gäbler
- GEOMAR Helmholtz-Zentrum für Ozeanforschung Kiel, 24148 Kiel, Germany; (A.E.); (R.G.)
| | - Carsten Schultz
- Kiel Institute for Responsible Innovation, Chair of Technology Management, Kiel University (Christian-Albrechts-Universität zu Kiel), 24118 Kiel, Germany; (M.Z.); (T.M.)
| |
Collapse
|
34
|
Abdullah, Ahmad N, Xiao J, Tian W, Khan NU, Hussain M, Ahsan HM, Hamed YS, Zhong H, Guan R. Gingerols: Preparation, encapsulation, and bioactivities focusing gut microbiome modulation and attenuation of disease symptoms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156352. [PMID: 39740381 DOI: 10.1016/j.phymed.2024.156352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/17/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut dysbiosis, chronic diseases, and microbial recurrent infections concerns have driven the researchers to explore phytochemicals from medicinal and food homologous plants to modulate gut microbiota, mitigate diseases, and inhibit pathogens. Gingerols have attracted attention as therapeutic agents due to their diverse biological activities like gut microbiome regulation, gastro-protective, anti-inflammatory, anti-microbial, and anti-oxidative effects. PURPOSE This review aimed to summarize the gingerols health-promoting potential, specifically focusing on the regulation of gut microbiome, attenuation of disease symptoms, mechanisms of action, and signaling pathways involved. METHOD Research findings from experimental and clinical studies have been summarized regarding gingerols effects on the modulation of gut microbiome and its metabolites, and attenuation of disease symptoms. RESULTS Gingerols are phenolic compounds characterized by a common 3-methoxy-4-hydroxyphenyl moiety in their chemical structures, and further divided into different gingerol types, including gingerols (major), shogaols, paradols, gingerdiols, gingerdiones, and zingerones (minor). Advanced extraction techniques (e.g., ionic liquid-based-, enzyme-assisted-, microwave-assisted-, pressurized liquid-, ultrasound-assisted-, and supercritical fluid extractions) were reported as optimal alternatives to conventional methods for gingerols extraction. Research studies reported that gingerols positively modulated the composition of gut microbiome that helped to combat disease symptoms (e.g., obesity by decreasing weight gain- (Lactobacillus reuteri and Lachnospiraceae) and increasing weight loss associated-bacteria (Akkermansia, Muribaculaceae, and Alloprevotella). Gingerols intervention also ameliorated ulcerative colitis by increasing relative abundance of the beneficial bacteria (Akkermansia, Lachnospiraceae NK4A136, and Muribaculaceae_norank), and decreasing pathogenic microorganisms (Bacteroides, Parabacteroides, and Desulfovibrio). Emerging delivery systems (e.g., microcapsules, nanoparticles, nanostructured lipid carriers, nanoemulsions, and nanoliposomes) can enhance the bioavailability and therapeutic efficacy of gingerols by preserving their inherent properties and addressing challenges of stability, solubility, and absorption. CONCLUSION Gingerols are promising therapeutic agents to modulate gut microbiome (increase beneficial bacteria and inhibit pathogenic microbes), and attenuate chronic disease symptoms such as diabetes, colitis, obesity, oxidative stress, and cancer. Despite significant progress, challenges persist in transforming research findings into industrial applications, such as stability and solubility during processing and low bioavailability in the distal gut to impart desirable health benefits.
Collapse
Affiliation(s)
- Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Naveed Ahmad
- Multan College of Food & Nutrition Sciences, Multan Medical & Dental College, Multan, Pakistan
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenni Tian
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hafiz Muhammad Ahsan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yahya Saud Hamed
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
35
|
Abdulhakeem Mansour Alhasbary A, Hashimah Ahamed Hassain Malim N, Zuraidah Mohamad Zobir S. Exploring natural products potential: A similarity-based target prediction tool for natural products. Comput Biol Med 2025; 184:109351. [PMID: 39536385 DOI: 10.1016/j.compbiomed.2024.109351] [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: 02/08/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Natural products are invaluable resources in drug discovery due to their substantial structural diversity. However, predicting their interactions with druggable protein targets remains a challenge, primarily due to the limited availability of bioactivity data. This study introduces CTAPred (Compound-Target Activity Prediction), an open-source command-line tool designed to predict potential protein targets for natural products. CTAPred employs a two-stage approach, combining fingerprinting and similarity-based search techniques to identify likely drug targets for these bioactive compounds. Despite its simplicity, the tool's performance is comparable to that of more complex methods, demonstrating proficiency in target retrieval for natural product compounds. Furthermore, this study explores the optimal number of reference compounds most similar to the query compound, aiming to refine target prediction accuracy. The findings demonstrated the superior performance of considering only the most similar reference compounds for target prediction. CTAPred is freely available at https://github.com/Alhasbary/CTAPred, offering a valuable resource for deciphering natural product-target associations and advancing drug discovery.
Collapse
Affiliation(s)
| | | | - Siti Zuraidah Mohamad Zobir
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), National Institutes of Biotechnology Malaysia (NIBM), Halaman Bukit Gambir, 11700, Gelugor, Pulau Pinang, Malaysia.
| |
Collapse
|
36
|
Li Y, Wei J, Ye Z, Ji C, Li W, Xu L, Zhou Z. Essential Oils from Citrus Peels Promote Calcium Overload-Induced Calcicoptosis in U251 Cells. Antioxidants (Basel) 2024; 14:11. [PMID: 39857347 PMCID: PMC11762846 DOI: 10.3390/antiox14010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Citrus peel essential oils (CPEOs) have demonstrated substantial medicinal potential for glioblastoma treatment because of their extensive antitumor effects, low potential for drug resistance, and ability to cross the human blood-brain barrier. In this study, the chemical compositions of five CPEOs were analyzed via gas chromatography-mass spectrometry (GC-MS). CCK8 assays were used to evaluate the ability of five CPEOs to inhibit U251 human glioblastoma cells, and XLB and RA were selected for further investigation. Through wound healing assays and cell cycle and apoptosis analyses via flow cytometry, it was revealed that these CPEOs inhibited cell migration, arrested the cell cycle at G1/G0, and induced apoptosis with similar levels of inhibition. After CPEOs treatment, the intracellular Ca2+ content and reactive oxygen species levels in U251 cells increased significantly, whereas the mitochondrial membrane potential decreased. Additionally, the antioxidant enzyme system (SOD, POD, CAT, and GR) and the nonenzymatic defense system (GSH) were inhibited, leading to an increase in lipid peroxidation. qRT-PCR indicated the significant upregulation of intracellular calcium ion signaling pathways and the upregulation of mitochondrial apoptosis-related genes. Additionally, the activation of calcicoptosis-related indicators induced by the CPEOs could be reversed by inhibitor treatment, confirming that both of the selected CPEOs inhibit tumors by activating calcicoptosis-related pathways. These findings highlight the immense potential of CPEOs in healthcare and pharmaceutical applications by not only providing a scientific basis for the potential application of CPEOs in the treatment of glioblastoma but also offering new insights for the development of novel antitumor drugs.
Collapse
Affiliation(s)
- Yurong Li
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Juanjuan Wei
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Zimao Ye
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Chen Ji
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Wenji Li
- School of Design, Chongqing Industry Polytechnic College, Chongqing 401120, China;
| | - Li Xu
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Beibei District, Chongqing 400715, China
| | - Zhiqin Zhou
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
- Southwest Institute of Fruits Nutrition, Banan District, Chongqing 400054, China
| |
Collapse
|
37
|
Han C, Song A, He Y, Yang L, Chen L, Dai W, Wu Q, Yuan S. Genome mining and biosynthetic pathways of marine-derived fungal bioactive natural products. Front Microbiol 2024; 15:1520446. [PMID: 39726967 PMCID: PMC11669671 DOI: 10.3389/fmicb.2024.1520446] [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: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Marine fungal natural products (MFNPs) are a vital source of pharmaceuticals, primarily synthesized by relevant biosynthetic gene clusters (BGCs). However, many of these BGCs remain silent under standard laboratory culture conditions, delaying the development of novel drugs from MFNPs to some extent. This review highlights recent efforts in genome mining and biosynthetic pathways of bioactive natural products from marine fungi, focusing on methods such as bioinformatics analysis, gene knockout, and heterologous expression to identify relevant BGCs and elucidate the biosynthetic pathways and enzyme functions of MFNPs. The research efforts presented in this review provide essential insights for future gene-guided mining and biosynthetic pathway analysis in MFNPs.
Collapse
Affiliation(s)
- Caihua Han
- School of Bioengineering, Zunyi Medical University, Zhuhai, China
| | - Anjing Song
- School of Bioengineering, Zunyi Medical University, Zhuhai, China
| | - Yueying He
- School of Bioengineering, Zunyi Medical University, Zhuhai, China
| | - Liu Yang
- School of Bioengineering, Zunyi Medical University, Zhuhai, China
| | - Litong Chen
- Center of Ocean Expedition, School of Atmospheric Science, Sun Yat-sen University, Zhuhai, China
| | - Wei Dai
- Teaching and Experimental Center, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qilin Wu
- School of Bioengineering, Zunyi Medical University, Zhuhai, China
| | - Siwen Yuan
- School of Bioengineering, Zunyi Medical University, Zhuhai, China
| |
Collapse
|
38
|
Chen H, Lu D, Xiao Z, Li S, Zhang W, Luan X, Zhang W, Zheng G. Comprehensive applications of the artificial intelligence technology in new drug research and development. Health Inf Sci Syst 2024; 12:41. [PMID: 39130617 PMCID: PMC11310389 DOI: 10.1007/s13755-024-00300-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 07/27/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose Target-based strategy is a prevalent means of drug research and development (R&D), since targets provide effector molecules of drug action and offer the foundation of pharmacological investigation. Recently, the artificial intelligence (AI) technology has been utilized in various stages of drug R&D, where AI-assisted experimental methods show higher efficiency than sole experimental ones. It is a critical need to give a comprehensive review of AI applications in drug R &D for biopharmaceutical field. Methods Relevant literatures about AI-assisted drug R&D were collected from the public databases (Including Google Scholar, Web of Science, PubMed, IEEE Xplore Digital Library, Springer, and ScienceDirect) through a keyword searching strategy with the following terms [("Artificial Intelligence" OR "Knowledge Graph" OR "Machine Learning") AND ("Drug Target Identification" OR "New Drug Development")]. Results In this review, we first introduced common strategies and novel trends of drug R&D, followed by characteristic description of AI algorithms widely used in drug R&D. Subsequently, we depicted detailed applications of AI algorithms in target identification, lead compound identification and optimization, drug repurposing, and drug analytical platform construction. Finally, we discussed the challenges and prospects of AI-assisted methods for drug discovery. Conclusion Collectively, this review provides comprehensive overview of AI applications in drug R&D and presents future perspectives for biopharmaceutical field, which may promote the development of drug industry.
Collapse
Affiliation(s)
- Hongyu Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyi Xiao
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Shensuo Li
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangyong Zheng
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
39
|
Jing QD, A JD, Liu LX, Fan HN. Current status of drug therapy for alveolar echinococcosis. World J Hepatol 2024; 16:1243-1254. [PMID: 39606163 PMCID: PMC11586754 DOI: 10.4254/wjh.v16.i11.1243] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024] Open
Abstract
Alveolar echinococcosis (AE) is a chronic zoonotic parasitic disease caused by infection with Echinococcus multilocularis. AE is associated with a high mortality rate and poses a significant threat to human health. The primary treatment for AE is surgical resection of the lesions; however, owing to its long incubation period and insidious disease progression, many patients are diagnosed only after the onset of complications such as liver cirrhosis, jaundice, and portal hypertension, which preclude curative surgical intervention. For patients who are unwilling or unable to undergo surgery, lifelong administration of anti-AE medications is necessary. Benzimidazole compounds, such as albendazole and mebendazole, are the current mainstays of treatment, offering good efficacy. Nevertheless, these medications primarily inhibit parasite proliferation rather than eradicate the infection, and their long-term use can lead to significant drug-related toxic effects. Consequently, there is an urgent need to develop new therapeutic strategies that convey better efficacy and reduce the adverse effects associated with current treatments. Recent advancements in AE therapy include novel synthetic compounds such as antiviral agents, antibiotics, antineoplastic agents, immunosuppressants, and antiangiogenic agents, as well as natural compounds derived from traditional Chinese and Tibetan medicine. These new drugs show promising clinical potential because they interfere with parasitic metabolic pathways and cellular structures. This review aims to discuss recent research on AE drug therapy, including mechanisms of action, dosing regimens, signalling pathways, and therapeutic outcomes, with a goal of providing new insights and directions for the development of anti-AE drugs and summarizing current advancements in AE pharmacotherapy.
Collapse
Affiliation(s)
- Qin-Dong Jing
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining 810000, Qinghai Province, China
- School of Clinical Medicine, Qinghai University, Xining 810000, Qinghai Province, China
| | - Ji-De A
- Department of Hepatic Hydatidosis, Qinghai Provincial People's Hospital, Xining 810007, Qinghai Province, China
| | - Lin-Xun Liu
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining 810000, Qinghai Province, China
| | - Hai-Ning Fan
- Department of Hepatobiliary and Pancreatic Surgery, Qinghai Province Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810001, Qinghai Province, China.
| |
Collapse
|
40
|
Perrier F, Morice J, Gueulle S, Géry A, Riboulet-Bisson E, Garon D, Muller C, Desriac F. Assessing Normandy Soil Microbial Diversity for Antibacterial Activities Using Traditional Culture and iChip Methods. Microorganisms 2024; 12:2422. [PMID: 39770625 PMCID: PMC11679952 DOI: 10.3390/microorganisms12122422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
Uncultured microorganisms represent a promising and untapped source of antibacterial compounds, crucial in the fight against the significant threat of antimicrobial resistance (AMR). In this study, both traditional and isolation chip (iChip) cultivation techniques were employed to enhance the recovery of known and unknown microorganisms from soils located in Normandy, France. The isolates obtained were identified using 16S rDNA or ITS regions analysis and MALDI-TOF mass spectrometry and were screened for antibacterial activity. A total of 386 isolates, belonging to 6 microbial phyla and distributed across 65 genera, were recovered using both methods. In total, 11 isolates are potentially new bacterial species, and 34 were associated with 22 species described recently. The iChip method yielded a higher diversity of microorganisms (47 genera) than the traditional method (38 genera) and was particularly effective in enriching Actinomycetota. Antibacterial screening against target bacteria showed that 85 isolates (22%) exhibited antibacterial activity. The Streptomyces, Pseudomonas, and Bacillaceae taxa accounted for most antibacterial-producing bacteria with some presenting promising undescribed characteristics. Other active isolates were affiliated with less-known antibacterial producers such as Arthrobacter, Chryseobacterium, Delftia, Ensifer, Flavobacterium, Rahnella, and Stenotrophomonas, among others. These results highlight the potential of our microbial collection as a source of new antibacterial natural products.
Collapse
Affiliation(s)
- Fabien Perrier
- Université de Caen Normandie, CBSA UR 4312, UFR des Sciences, Campus 1, F-14000 Caen, France; (J.M.); (S.G.); (E.R.-B.); (C.M.)
| | - Juliette Morice
- Université de Caen Normandie, CBSA UR 4312, UFR des Sciences, Campus 1, F-14000 Caen, France; (J.M.); (S.G.); (E.R.-B.); (C.M.)
| | - Sabrina Gueulle
- Université de Caen Normandie, CBSA UR 4312, UFR des Sciences, Campus 1, F-14000 Caen, France; (J.M.); (S.G.); (E.R.-B.); (C.M.)
| | - Antoine Géry
- Université de Caen Normandie, ToxEMAC-ABTE UR 4651, UFR des Sciences, Campus 1, F-14000 Caen, France; (A.G.); (D.G.)
| | - Eliette Riboulet-Bisson
- Université de Caen Normandie, CBSA UR 4312, UFR des Sciences, Campus 1, F-14000 Caen, France; (J.M.); (S.G.); (E.R.-B.); (C.M.)
| | - David Garon
- Université de Caen Normandie, ToxEMAC-ABTE UR 4651, UFR des Sciences, Campus 1, F-14000 Caen, France; (A.G.); (D.G.)
| | - Cécile Muller
- Université de Caen Normandie, CBSA UR 4312, UFR des Sciences, Campus 1, F-14000 Caen, France; (J.M.); (S.G.); (E.R.-B.); (C.M.)
| | - Florie Desriac
- Université de Caen Normandie, CBSA UR 4312, UFR des Sciences, Campus 1, F-14000 Caen, France; (J.M.); (S.G.); (E.R.-B.); (C.M.)
| |
Collapse
|
41
|
Calzoni E, Bertoldi A, Cesaretti A, Alabed HBR, Cerrotti G, Pellegrino RM, Buratta S, Urbanelli L, Emiliani C. Aloe Extracellular Vesicles as Carriers of Photoinducible Metabolites Exhibiting Cellular Phototoxicity. Cells 2024; 13:1845. [PMID: 39594594 PMCID: PMC11592872 DOI: 10.3390/cells13221845] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The growing interest in plant-origin active molecules with medicinal properties has led to a revaluation of plants in the pharmaceutical field. Plant-derived extracellular vesicles (PDEVs) have emerged as promising candidates for next-generation drug delivery systems due to their ability to concentrate and deliver a plethora of bioactive molecules. These bilayer membranous vesicles, whose diameter ranges from 30 to 1000 nm, are released by different cell types and play a crucial role in cross-kingdom communication between plants and humans. Notably, PDEVs have demonstrated efficacy in treating various diseases, including cancer, alcoholic liver disease, and inflammatory bowel disease. However, further research on plant vesicles is necessary to fully understand their traits and purposes. This study investigates the phototoxic effects of extracellular vesicles (EVs) from Aloe arborescens, Aloe barbadensis, and Aloe chinensis on the human melanoma cell line SK-MEL-5, focusing on their anthraquinone content, recognized as natural photosensitizers. The phototoxic impact of Aloe EVs is associated with ROS production, leading to significant oxidative stress in melanoma cells, as validated by a metabolome analysis. These findings suggest that EVs from Aloe arborescens, Aloe barbadensis, and Aloe chinensis hold promise as potential photosensitizers, thus highlighting their potential for future application in photodynamic cancer therapy and providing valuable insights into the possible utilization of PDEVs for therapeutic purposes.
Collapse
Affiliation(s)
- Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Agnese Bertoldi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Alessio Cesaretti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
- Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
- Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| |
Collapse
|
42
|
Wen Y, Li Y, Li BB, Liu P, Qiu M, Li Z, Xu J, Bi B, Zhang S, Deng X, Liu K, Zhou S, Wang Q, Zhao J. Pyroptosis induced by natural products and their derivatives for cancer therapy. Biomater Sci 2024; 12:5656-5679. [PMID: 39429101 DOI: 10.1039/d4bm01023j] [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: 10/22/2024]
Abstract
Natural products, which are compounds extracted and/or refined from plants and microbes in nature, have great potential for the discovery of therapeutic agents, especially for infectious diseases and cancer. In recent years, natural products have been reported to induce multiple cell death pathways to exhibit antitumor effects. Among them, pyroptosis is a unique programmed cell death (PCD) characterized by continuous cell membrane permeability and intracellular content leakage. According to the canonical and noncanonical pathways, the formation of gasdermin-N pores involves a variety of transcriptional targets and post-translational modifications. Thus, tailored control of PCD may facilitate dying cells with sufficient immunogenicity to activate the immune system to eliminate other tumor cells. Therefore, we summarized the currently reported natural products or their derivatives and their nano-drugs that induce pyroptosis-related signaling pathways. We reviewed six main categories of bioactive compounds extracted from natural products, including flavonoids, terpenoids, polyphenols, quinones, artemisinins, and alkaloids. Correspondingly, the underlying mechanisms of how these compounds and their derivatives engage in pyroptosis are also discussed. Moreover, the synergistic effect of natural bioactive compounds with other antitumor therapies is proposed as a novel therapeutic strategy for traditional chemotherapy, radiotherapy, chemodynamic therapy, photodynamic therapy, photothermal therapy, hyperthermal therapy, and sonodynamic therapy. Consequently, we provide insights into natural products to develop a novel antitumor therapy or qualified adjuvant agents by inducing pyroptosis, which may eventually be applied clinically.
Collapse
Affiliation(s)
- Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Bin-Bin Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Peng Liu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Miaojuan Qiu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Zihang Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Jiaqi Xu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Bo Bi
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Xinyi Deng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Kaiyuan Liu
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shangbo Zhou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Qiang Wang
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
43
|
Ibrahim MAA, Ali SSM, Abdelrahman AHM, Abdeljawaad KAA, Sidhom PA, Sayed SRM, El-Tayeb MA, Paré PW, Hegazy MEF. Naturally Occurring Plant-Based Anticancerous Candidates as Potential ERK2 Inhibitors: In-Silico Database Mining and Molecular Dynamics Simulations. Chem Biodivers 2024; 21:e202401238. [PMID: 39075025 DOI: 10.1002/cbdv.202401238] [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: 05/15/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024]
Abstract
The evolutionarily conserved extracellular signal-regulated kinase 2 (ERK2) is involved in regulating cellular signaling in both normal and pathological conditions. ERK2 expression is critical for human development, while hyperactivation is a major factor in tumor progression. Up to now, there have been no approved inhibitors that target ERK2, and as such, here we report on screening of a naturally occurring plant-based anticancerous compound-activity-target (NPACT) database for prospective ERK2 inhibitors. More than 1,500 phytochemicals were screened using in-silico molecular docking and molecular dynamics (MD) approaches. NPACT compounds with a docking score lower than a co-crystallized LHZ inhibitor (calc. -10.5 kcal/mol) were subjected to MD simulations. Binding energies (ΔGbinding) of inhibitor-ERK2 complexes over the MD course were estimated using an MM-GBSA approach. Based on MM-GBSA//100 ns MD simulations, the steroid zhankuic acid C (NPACT01034) demonstrated greater binding affinity against ERK2 protein than LHZ, with ΔGbinding values of -50.0 and -47.7 kcal/mol, respectively. Structural and energetical analyses throughout the MD course demonstrated stabilization of zhankuic acid C complexed with ERK2 protein. The anticipated ADMET properties of zhankuic acid C indicated minimal toxicity. Moreover, in-silico evaluation of fourteen ERK2 inhibitors in clinical trials demonstrated the higher binding affinity of zhankuic acid C towards ERK2 protein.
Collapse
Affiliation(s)
- Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
- School of Health Sciences, Westville Campus, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Sara S M Ali
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Khlood A A Abdeljawaad
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Shaban R M Sayed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mohamed A El-Tayeb
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Paul W Paré
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, TX, 79409, USA
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, 55128, Germany
| |
Collapse
|
44
|
Liu X, Li R, Zhou X. Structurally diverse indole alkaloids with cytotoxicity from Lonicera Japonica-associated endophytic fungus Penicillium ochrochloron YT2022-65. Nat Prod Res 2024; 38:4071-4075. [PMID: 37867305 DOI: 10.1080/14786419.2023.2272778] [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: 07/30/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 10/24/2023]
Abstract
Natural products, especially fungal secondary metabolites, have been served as valuable sources of drug leads in pharmaceutical industry. Medicinal plants-associated endophytic fungi possess a well-developed secondary metabolism. In this study, chemical investigation on Penicillium ochrochloron YT2022-65, an endophytic fungus associated with Lonicera Japonica, led to the isolation of six structurally diversified indole alkaloids, including a new one, namely peniochroloid A (1), as well as five previously reported alkaloids, flavonoid B (2), brocaeloid C (3), isoroquefortine C (4), roquefortine C (5), and dihydrocarneamide A (6). Their structures, including the absolute configuration of 1, were determined by a combined analysis of HRESIMS, NMR spectroscopic data, and calculation of the optical rotation. Their cytotoxicity against A549, HepG2, MCF-7, and THP-1 cell lines were evaluated in vitro. The new compound 1 was found to possess considerable cytotoxicity against MCF-7 and THP-1 cell lines with IC50 values of 10.2 and 11.0 μM, respectively.
Collapse
Affiliation(s)
- Xingjie Liu
- Medicine & Pharmacy Research Center, Binzhou Medical University, Yantai, China
| | - Ruonan Li
- Medicine & Pharmacy Research Center, Binzhou Medical University, Yantai, China
| | - Xiaofei Zhou
- Community Health Service center of Luoyang Road, Qingdao, China
| |
Collapse
|
45
|
Wang S, Zhang Y, Yu R, Chai Y, Liu R, Yu J, Qu Z, Zhang W, Zhuang C. Labeled and Label-Free Target Identifications of Natural Products. J Med Chem 2024; 67:17980-17996. [PMID: 39360958 DOI: 10.1021/acs.jmedchem.4c01576] [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: 10/25/2024]
Abstract
Target identification, employing chemical proteomics, constitutes a continuous challenging endeavor in the drug development of natural products (NPs). Understanding their targets is crucial for deciphering their mechanisms and developing potential probes or drugs. Identifications fall into two main categories: labeled and label-free techniques. Labeled methods use the molecules tagged with markers such as biotin or fluorescent labels to easily detect interactions with target proteins. Thorough structure-activity relationships are essential before labeling to avoid changes in the biological activity or binding specificity. In contrast, label-free technologies identify target proteins without modifying natural products, relying on changes in the stability, thermal properties, or precipitation in the presence or absence of these products. Each approach has its advantages and disadvantages, offering a comprehensive understanding of the mechanisms and therapeutic potential of the NPs. Here, we summarize target identification techniques for natural molecules, highlight case studies of notable NPs, and explore future applications and directions.
Collapse
Affiliation(s)
- Shuyu Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Yu Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Ruizhi Yu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yue Chai
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ruyun Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Zhuo Qu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
46
|
de Souza TA, Pereira LHA, Alves AF, Dourado D, Lins JDS, Scotti MT, Scotti L, Abreu LS, Tavares JF, Silva MS. Jatropha Diterpenes: An Updated Review Concerning Their Structural Diversity, Therapeutic Performance, and Future Pharmaceutical Applications. Pharmaceuticals (Basel) 2024; 17:1399. [PMID: 39459038 PMCID: PMC11510188 DOI: 10.3390/ph17101399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The Euphorbiaceae family is a rich source of bioactive terpenoids. Among its genera, Jatropha is a conspicuous producer of diterpenes and includes approximately 175 species, many of which have medicinal uses. To date, 140 diterpenes from Jatropha (JTDs) have been reported. Given their structural diversity and notable biological activities, this work aims to highlight the pharmaceutical potential of JTDs. To achieve this goal, an extensive literature review was conducted, encompassing studies on structural elucidation through NMR and pharmacological assays, both in vitro and in vivo. Based on 132 selected papers, a thorough discussion is presented on the biosynthesis, extraction, isolation, and structural characterization of JTDs, including a compilation of their 13C NMR chemical shifts. The review also covers their synthetic production and biological effects. Additionally, an in silico analysis predicting the drug-likeness of 141 JTDs was carried out. Notably, the occurrence of macrocyclic diterpenes has doubled in the past decade, and the summary of their NMR data provides a useful resource for future research. Furthermore, 21 distinct pharmacological activities were identified, with potent cytotoxic effects targeting new molecular pathways being particularly significant. Recent advances highlight the contributions of modern approaches in organic synthesis and the pharmacological evaluation of natural products. The drug-likeness analysis identified JTD classes and compounds with favorable physicochemical and ADMET features for pharmaceutical development. In light of these findings, the use of nanotechnology is proposed as a future direction for continued research on JTDs, a fascinating class of natural compounds. This work opens up new avenues for the study of Euphorbiaceae species, particularly the Jatropha genus and its bioactive compounds.
Collapse
Affiliation(s)
- Thalisson A. de Souza
- Multi-User Characterization and Analysis Laboratory, Research Institute for Drugs and Medicines (IpeFarM), Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (T.A.d.S.); (L.H.A.P.); (J.d.S.L.); (J.F.T.)
| | - Luiz H. A. Pereira
- Multi-User Characterization and Analysis Laboratory, Research Institute for Drugs and Medicines (IpeFarM), Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (T.A.d.S.); (L.H.A.P.); (J.d.S.L.); (J.F.T.)
| | - Alan F. Alves
- Laboratory of Cheminformatics, Program of Post-Graduation on Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (A.F.A.); (M.T.S.); (L.S.)
| | - Douglas Dourado
- Department of Immunology, Instituto Aggeu Magalhães, Fiocruz, Recife 50670-420, Brazil;
| | - Jociano da S. Lins
- Multi-User Characterization and Analysis Laboratory, Research Institute for Drugs and Medicines (IpeFarM), Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (T.A.d.S.); (L.H.A.P.); (J.d.S.L.); (J.F.T.)
| | - Marcus T. Scotti
- Laboratory of Cheminformatics, Program of Post-Graduation on Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (A.F.A.); (M.T.S.); (L.S.)
| | - Luciana Scotti
- Laboratory of Cheminformatics, Program of Post-Graduation on Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (A.F.A.); (M.T.S.); (L.S.)
| | - Lucas S. Abreu
- Department of Organic Chemistry, Universidade Federal Fluminense, Niterói 24220-900, Brazil;
| | - Josean F. Tavares
- Multi-User Characterization and Analysis Laboratory, Research Institute for Drugs and Medicines (IpeFarM), Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (T.A.d.S.); (L.H.A.P.); (J.d.S.L.); (J.F.T.)
| | - Marcelo S. Silva
- Multi-User Characterization and Analysis Laboratory, Research Institute for Drugs and Medicines (IpeFarM), Universidade Federal da Paraíba, João Pessoa 58051-900, Brazil; (T.A.d.S.); (L.H.A.P.); (J.d.S.L.); (J.F.T.)
| |
Collapse
|
47
|
Venable KE, Lee CC, Francis J. Addressing Mental Health in Rural Settings: A Narrative Review of Blueberry Supplementation as a Natural Intervention. Nutrients 2024; 16:3539. [PMID: 39458533 PMCID: PMC11510281 DOI: 10.3390/nu16203539] [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: 08/14/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Depression and anxiety are major public health issues; however, there is an unmet need for novel, effective, and accessible treatments, particularly in rural communities. Blueberries are an unexplored nutraceutical for these conditions due to their excellent nutritional profile, with particularly high levels of polyphenols and anthocyanins and benefits on mood, cognition, and health. Here, we present a narrative review of the literature concerning the etiology and treatments of major depressive disorder (MDD) and generalized anxiety disorder (GAD). In both animal and human studies, blueberry supplementation can ameliorate behavioral symptoms of both anxiety and depression. The mechanistic underpinnings of these behavioral improvements are not fully defined, but likely involve biochemical alterations in the gut-brain axis, including to inflammatory cytokines, reactive oxygen species, and growth factors. We also review the limitations of traditional therapies in rural settings. Finally, we assess the potential benefit of nutraceutical interventions, particularly blueberries, as novel therapeutics for these distinct, yet related mental health issues.
Collapse
Affiliation(s)
- Katy E. Venable
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (C.C.L.); (J.F.)
| | | | | |
Collapse
|
48
|
Arora M, Singh AK, Kumar A, Singh H, Pathak P, Grishina M, Yadav JP, Verma A, Kumar P. Semisynthetic phytochemicals in cancer treatment: a medicinal chemistry perspective. RSC Med Chem 2024; 15:3345-3370. [PMID: 39430100 PMCID: PMC11484407 DOI: 10.1039/d4md00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer is the uncontrolled proliferation of abnormal cells that invade other areas, spread to other organs, and cause metastases, which is the most common cause of death. A review of all FDA-approved new molecular entities (NMEs) shows that natural products and derivatives account for over one-third of all NMEs. Before 1940, unmodified products and derivatives accounted for 43% and 14% of NME registrations, respectively. Since then, the share of unmodified products has decreased to 9.5% of all approved NMEs, while the share of derivatives has increased to 28%. Since the 1940s, semi-synthetic and synthetic derivatives of natural substances have gained importance, and this trend continues to date. In this study, we have discussed in detail isolated phytoconstituents with chemical modifications that are either FDA-approved or under clinical trials, such as podophyllotoxin, Taxol (paclitaxel, docetaxel), vinca alkaloids (vincristine, vinblastine), camptothecin, genistein, cephalotaxine, rohitukine, and many more, which may act as essential leads to the development of novel anticancer agents. Furthermore, we have also discussed recent developments in the most potent semisynthetic phytoconstituents, their unique properties, and their importance in cancer treatment.
Collapse
Affiliation(s)
- Meghna Arora
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Prateek Pathak
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, School of Pharmacy, GITAM (Deemed to be University) Hyderabad Campus India
| | - Maria Grishina
- Laboratory of Computational Modeling of Drugs, Higher Medical and Biological School, South Ural State University Chelyabinsk 454008 Russia
| | - Jagat Pal Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University Kanpur 209217 India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| |
Collapse
|
49
|
Unver T, Gurhan I. Unveiling the Chemical Constituents and Inhibitory Roles of Extracts from Pinus Pinea L. Nut and Nutshell: A Novel Source for Pharmaceutical Antimicrobials. Chem Biodivers 2024; 21:e202401208. [PMID: 39178285 DOI: 10.1002/cbdv.202401208] [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: 05/10/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/25/2024]
Abstract
Antibiotic resistance in infectious diseases has been a serious problem for the last century, and scientists have focused on discovering new natural antimicrobial agents. Pinus pinea has been used as a natural pharmacotherapeutic agent with antimutagenic, anticarcinogenic, and high antioxidant properties. In this study, GC-MS and LC-HR/MS were employed to analyze Pinus pinea L. nut and nutshell extracts. DPPH radical scavenging assay was performed to analyze the antioxidant properties of the extracts, but no activity was determined. GC-MS analysis showed that linoleic, oleic, and palmitic acids were the three most dominant fatty acids in nut and nutshell extracts, with ratios between 6.75 % and 47.06 % (v/v). LC-HR/MS revealed that the nutshell methanol extract had a higher phenolic content than other extracts, with vanillic acid (1.4071 mg/g). Antimicrobial activity assays showed that the minimum inhibitory concentrations (MIC) of the extracts varied between 5.94 and 190 mg/mL, and the most significant inhibition was seen in the nutshell methanol extract (MICs: between 5.94 and 47.5 mg/mL). Consequently, the antimicrobial activity of the extracts can be attributed to the dense fatty acids they contain, and the nutshell methanol extract showed the most potent inhibition related to the abundance of phenolic compounds in the extract.
Collapse
Affiliation(s)
- Tuba Unver
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Inonu University, 44280, Malatya, Turkey
| | - Ismet Gurhan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Inonu University, 44280, Malatya, Turkey
| |
Collapse
|
50
|
Banu HS, Parvin IS, Priyadharshini SD, Gayathiri E, Prakash P, Pratheep T. Molecular insights into the antioxidant and anticancer properties: A comprehensive analysis through molecular modeling, docking, and dynamics studies. J Cell Biochem 2024; 125:e30564. [PMID: 38747366 DOI: 10.1002/jcb.30564] [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: 12/07/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 10/13/2024]
Abstract
Plants are rich sources of therapeutic compounds that often lack the side effects commonly found in synthetic chemicals. Researchers have effectively synthesized pharmaceuticals from natural sources, taking inspiration from traditional medicine, in their pursuit of modern drugs. This study aims to evaluate the phenolic and flavonoid content of Solanum virginianum seeds using different solvent extracts, enzymatic assays including 2,2-diphenyl-1-picrylhydrazyl activity, reducing power, and superoxide activity. Our phytochemical screening identified active compounds, such as phenols, flavonoids, tannins, and alkaloids. The methanol extract notably possesses higher levels of total phenolic and flavonoid content in comparison to the other extracts. The results highlight the superior antioxidant activity of methanol-extracted leaves, demonstrated by their exceptional IC50 values, which surpass the established standard. In this study, molecular docking techniques were used to assess the binding affinity and to predict the binding conformation of the compounds. Quercetin 3-O beta-d-galactopyranoside displayed a binding energy of -8.35 kcal/mol with several important amino acid residues, PHE222, TRP440, ILE184, LEU192, VAL221, LEU218, SER185, and ALA188. Kaempferol 3-O-beta-l-glucopyranoside exhibited a binding energy of -8.33 kcal/mol, interacting with specific amino acid residues including ALA 441, VAL318, VAL322, MET307, ILI409, GLY442, and PHE439. The results indicate that the methanol extract has a distinct composition of biologically active constituents compared to the other extracts. Overall, seeds exhibit promise as natural antioxidants and potential agents for combating cancer. This study highlights the significance of utilizing the therapeutic capabilities of natural compounds and enhancing our comprehension of their pharmacological characteristics.
Collapse
Affiliation(s)
- Hamza Serina Banu
- Department of Chemistry, Vellalar College for Women (Autonomous), Erode, Tamilnadu, India
| | - Ismail Sheriff Parvin
- Department of Chemistry, Vellalar College for Women (Autonomous), Erode, Tamilnadu, India
| | | | - Ekambaram Gayathiri
- Department of Plant Biology and Plant Biotechnology, Guru Nanak College (Autonomous), Chennai, India
| | - Palanisamy Prakash
- Department of Botany, Periyar University, Periyar Palkalai Nagar, Salem, Tamil Nadu, India
| | - Thangaraj Pratheep
- Department of Biotechnology, Rathinam College of Arts and Science, Coimbatore, Tamil Nadu, India
| |
Collapse
|