AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (34.6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access | Online First

Co-assembly strategies of natural plant compounds for improving their bioavailability

Yu-Hang MiaoXin WangXiao-Min ZhaoYan-Wei HuXin LiuDa-Wei Deng( )
Department of Biomedical Engineering, and Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
Show Author Information

Highlights

(1) Exploring the implications of traditional Chinese medicine decoction methods on the collaborative assembly strategy of natural plant compounds.

(2) Discussed the structural characteristics of flavonoids, triterpenes, polyphenols, phenylpropanoids, and alkaloid compounds, as well as typical collaborative assembly examples.

(3) Outlined the potential mechanisms of collaborative assembly of typical natural plant compounds.

Graphical Abstract

Co-assembly strategies effectively increase the bioavailability of natural plant compounds.

Abstract

Natural plant compounds have long served as crucial sources of medicinal therapies, spanning millennia and underpinning the field of drug discovery and development. Their profound impact is rooted in their extensive diversity and broad spectrum of pharmacological activities. Inspired by the decoction methods of traditional Chinese medicine, we have delved into the co-assembly strategy of natural plant compounds. This strategy involves the coalescence of active components from various drugs into nanocomplexes via non-covalent interactions during the decoction process. Consequently, this approach enhances the biocompatibility, biodegradability, and pharmacological efficacy of natural plant small molecules while mitigating their toxicity. The co-assembly strategy capitalizes on the inherent advantages of natural plant compounds, thereby expediting drug discovery by repurposing existing drugs. This review outlines the evolution of co-assembly strategies involving natural plant compounds, provides illustrative examples of co-assembly, elucidates the mechanisms underlying assembly, and highlights the resultant advantages. By shedding light on the transition from single molecule to multi-component interactions, this review advances our understanding, facilitating further research and the practical application of co-assembly strategies for natural plant compounds.

References

[1]

Mozaffarian, D., Blanck, H. M., Garfield, K. M., et al. A Food is Medicine approach to achieve nutrition security and improve health. Nature Medicine, 2022, 28: 2238–2240. https://doi.org/10.1038/s41591-022-02027-3

[2]

Zuo, W. F., Yao, L. P., Zhang, Y., et al. Gut microbiota: a magical multifunctional target regulated by medicine food homology species. Journal of Advanced Research, 2023, 52: 151–170. https://doi.org/10.1016/j.jare.2023.05.011

[3]

Tian, T. T., Luo, W. Y., Li, D., et al. The anti-aging mechanism of ginsenosides with medicine and food homology. Food & Function, 2023, 14: 9123. https://doi.org/10.1039/d3fo02580b

[4]

Su, Y., Bai, Q., Tao, H., et al. Prospects for the application of traditional Chinese medicine network pharmacology in food science research. Journal of the Science of Food and Agriculture, 2023, 103: 5183–5200. https://doi.org/10.1002/jsfa.12541

[5]
Venkatesan, P. Food is medicine: clinical trials show the health benefits of dietary interventions. Nature Medicine, 2024 , 30: 916-919. https://doi.org/10.1038/s41591-024-02891-1
[6]

Xiao, Y. L., Gong, Y., Qi, Y. J., et al. Effects of dietary intervention on human diseases: molecular mechanisms and therapeutic potential. Signal Transduction and Targeted Therapy, 2024, 9: 59. https://doi.org/10.1038/s41392-024-01771-x

[7]

Downer, S., Berkowitz, S. A., Harlan T. S., et al. Food is medicine: actions to integrate food and nutrition into healthcare. The BMJ, 2020, 369: m2482. https://doi.org/10.1136/bmj.m2482

[8]

Han, H. S., Koo, S. Y., Choi, K. Y., et al. Emerging nanoformulation strategies for phytocompounds and applications from drug delivery to phototherapy to imaging. Bioactive Materials, 2022, 14: 182–205. https://doi.org/10.1016/j.bioactmat.2021.11.027

[9]
Chopra, B., Dhingra, A. K. Natural products: a lead for drug discovery and development. Phytotherapy Research, 2021 , 35: 4660–4702. https://doi.org/10.1002/ptr.7099
[10]

Atanasov, A. G., Zotchev, S. B., Dirsch, V. M., et al. Natural products in drug discovery: advances and opportunities. Nature Reviews Drug Discovery, 2021, 20: 200–216. https://doi.org/10. 1038/s41573-020-00114-z

[11]
Zhang, L., Song, J. K., Kong, L. L., et al. The strategies and techniques of drug discovery from natural products. Pharmacology & Therapeutics, 2020 , 216: 107686. https://doi.org/10.1016/j.pharmthera.2020.107686
[12]

Haque, A., Amin, A. R. Perspectives on natural compounds in chemoprevention and treatment of cancer: an update with new promising compounds. European Journal of Cancer, 2021, 149: 165–183. https://doi.org/10.1016/j.ejca.2021.03.009

[13]
Zhuang, Y., Zhu, W., Chen, L. R., et al. Can the aggregation be a new approach for understanding the mechanism of Traditional Chinese Medicine? Journal of Ethnopharmacology, 2008 , 117: 378–384. https://doi.org/10.1016/j.jep.2008.02.017
[14]

Zhi, K. K., Wang, J. C., Zhao, H. T., et al. Self-assembled small molecule natural product gel for drug delivery: a breakthrough in new application of small molecule natural products. Acta Pharmaceutica Sinica B, 2020, 10: 913–927. https://doi.org/10.1016/j.apsb.2019.09.009

[15]

Wang, J. C., Li, X., Zhao, H. T., et al. A directed co-assembly of herbal small molecules into carrier-free nanodrugs for enhanced synergistic antitumor efficacy. Journal of Materials Chemistry B, 2021, 9: 1040. https://doi.org/10.1039/D0TB02071K

[16]

Hou, Y., Li, Q. L., Chen, M. Y., et al. Supramolecular assemblies based on natural small molecules: union would be effective. Materials Today Bio, 2022, 15: 100327. https://doi.org/10.1016/j.mtbio.2022.100327

[17]

Lin, R. Supramolecular nanostructures as drug carriers. Current Opinion in Chemical Engineering, 2015, 7: 75–83. https://doi.org/10.1016/j.coche.2014.11.005

[18]

Fu, S. Y., Yang, X. Recent advances in natural small molecules as drug delivery systems. Journal of Materials Chemistry B, 2023, 11: 4584. https://doi.org/10.1039/d3tb00070b

[19]

Cheng, J. J., Zhao, H. T., Li, B., et al. Photosensitive pro-drug nanoassemblies harboring a chemotherapeutic dormancy function potentiates cancer immunotherapy. Acta Pharmaceutica Sinica B, 2023, 13: 879–896. https://doi.org/10.1016/j.apsb.2022.06.008

[20]

Bag, B.G., Majumdar, R. Self-assembly of renewable nano-sized triterpenoids. The Chemical Record, 2017, 17: 841–873. https://doi.org/10.1002/tcr.201600123

[21]

Ma, W., Cheetham, A. G., Cui, H. G., et al. Building nanostructures with drugs. Nano Today, 2016, 11: 13–30. https://doi.org/10.1016/j.nantod.2015.11.003

[22]

Sun, X. Y., Zhang, H., Dong, M. N., et al. Green regenerative hydrogel wound dressing functionalized by natural drug-food homologous small molecule self-assembled nanospheres. Advanced Functional Materials, 2021, 32: 2106572. https://doi.org/10.1002/adfm.202106572

[23]

Zhao, R. R., Fan, L. L., Shen, Z. C., et al. Carrier-free nanodrug by co-assembly of chemotherapeutic agent and photosensitizer for cancer imaging and chemo-photo combination therapy. Acta Biomaterialia, 2018, 70: 197–210. https://doi.org/10.1016/j.actbio.2018.01.028

[24]

Huang, X. J., Jiang, X. Y., Wang, Z. L., et al. Co-assembled supramolecular hydrogel of salvianolic acid B and a phosphopeptide for enhanced wound healing. ACS Applied Materials & Interfaces, 2023, 15: 45606–45615. https://doi.org/10.1021/acsami.3c09219

[25]

Zheng, T. T., Wu, C. Y., Wang, J. R., et al. Fabrication of co-assembly from berberine and tannic acid for multidrug-resistant bacteria infection treatment. Pharmaceutics, 2023, 15: 1782. https://doi.org/10.3390/pharmaceutics15071782

[26]

Shen, N., Gan, Q., Liu, S., et al. Plant flavonoids: classification, distribution, biosynthesis, and antioxidant activity. Food Chemistry, 2022, 383: 132531. https://doi.org/10.1016/j.foodchem.2022.132531

[27]

Yin, Y., Liu, Y. H., Duan, M. W., et al. Optimizing the nucleic acid screening strategy to mitigate regional outbreaks of SARS-CoV-2 Omicron variant in China: a modeling study. Infectious Diseases of Poverty, 2023, 12: 1. https://doi.org/10.1186/s40249-022-01049-w

[28]

Xing, P., Zhong, Y. F., Cui, X., et al. Natural products in digestive tract tumors metabolism: functional and application prospects. Pharmacological Research, 2023, 191: 106766. https://doi.org/10.1016/j.phrs.2023.106766

[29]
Zhang, Y., Yu, C. H., Therapeutic potential of curcuma oil and its terpenoids in gynecological cancers. Biomedicine & Pharmacotherapy, 2023 , 157: 114016. https://doi.org/10.1016/j.biopha.2022.114016
[30]

Li, W., Yan, X. T., Yang, S. Y., et al. Coumarins and lignans from Zanthoxylum schinifolium and their anticancer activities. Journal of Agricultural and Food Chemistry, 2013, 61: 10730–10740. https://doi.org/10.1021/jf403479c

[31]

Huang, H. B., Wang, X. C., He, W. Y., et al. Self-Assembly of naturally small molecules into supramolecular fibrillar networks for wound healing. Advanced Healthcare Materials, 2022, 11: 2102476. https://doi.org/10.1002/adhm.202102476

[32]

Lia, Y., Lin, J. Y., Cai, Z. X., et al. Tumor microenvironment-activated self-recognizing nanodrug through directly tailored assembly of small-molecules for targeted synergistic chemotherapy. Journal of Controlled Release, 2020, 321: 222–235. https://doi.org/10.1016/j.jconrel.2020.02.025

[33]

Rybtchinski, B. Adaptive supramolecular nanomaterials based on strong noncovalent interactions. ACS Nano, 2011, 5: 6791–6818. https://doi.org/10.1021/nn2025397

[34]

Liu, D. L., Cai, Z. P., Li, Y., et al. Self-assembly of superstructures at all scales. Matter, 2021, 4: 927–941. https://doi.org/10.1016/j.matt.2020.12.020

[35]
Maleki, S. J., Crespo, J. F., Cabanillasb, B. Anti-inflammatory effects of flavonoids. Food Chemistry, 2019 , 299: 125124. https://doi.org/10.1016/j.foodchem.2019.125124
[36]

Sun, M., Zhao, C. L., Huang, J. X., et al. Herb-Functionalized chronic wound dressings for enhancing biological functions: multiple flavonoids coordination driven strategy. Advanced Functional Materials, 2022, 32: 2204291. https://doi.org/10.1002/adfm.202204291

[37]

Hui, Z., Wen, H., Zhu, J. L., et al. Discovery of plant-derived anti-tumor natural products: potential leads for anti-tumor drug discovery. Bioorganic Chemistry, 2024, 142: 106957. https://doi.org/10.1016/j.bioorg.2023.106957

[38]

Li, T., Wang, P. L., Guo, W. B., et al. Natural berberine-based Chinese herb medicine assembled nanostructures with modified antibacterial application. ACS Nano, 2019, 13: 6770–6781. https://doi.org/10.1021/acsnano.9b01346

[39]

He, J. Hydrophilic co-assemblies of two hydrophobic biomolecules improving the bioavailability of silybin. Food & Function, 2020, 11: 3910828. https://doi.org/10.1039/d0fo01882a

[40]
Heger, M., Broekgaarden, M., Michel, M. C., The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacological Reviews, 2014 , 66: 222–307. https://doi.org/10.1124/pr.110.004044
[41]

Dai, Y. M., Li, J. Y., Wang, M. F., et al. Co-assembly of curcumin and a cystine bridged peptide to construct tumor-responsive nano-micelles for efficient chemotherapy. Journal of Materials Chemistry, B, 2020, 8: 1944. https://doi.org/10.1039/c9tb02625h

[42]

Jia, Y. P., Zhan, W. J., Wang, Y., et al. Sustained release of neuroprotective drugs curcumin and edaravone from supramolecular hydrogel for ischemic stroke treatment. Advanced Functional Materials, 2023, 33: 2303930. https://doi.org/10.1002/adfm.202303930

[43]

Patra, S., Nayak, R., Behera, C., et al. Chemotherapeutic efficacy of curcumin and resveratrol against cancer: chemoprevention, chemoprotection, drug synergism and clinical pharmacokinetics. Seminars in Cancer Biology, 2021, 73: 310–320. https://doi.org/10.1016/j.semcancer.2020.10.010

[44]
Zou, P. R., Zhang, F., Thakur, K., Hydrophilic co-assembly of wheat gluten proteins and wheat bran cellulose improving the bioavailability of curcumin. Food Chemistry, 2022 , 397: 133807. https://doi.org/10.1016/j.foodchem.2022.133807
[45]

Lu, M. Q., Huang, Y. F., Yao, J. R., et al. Animal protein-plant protein composite nanospheres for dual-drug loading and synergistic cancer therapy. Journal of Materials Chemistry B, 2022, 10: 3798. https://doi.org/10.1039/D2TB00368F

[46]

Mao, L., PAN, Q. Y., Yuan, F., et al. Formation of soy protein isolate-carrageenan complex coacervates for improved viability of Bifidobacterium longum during pasteurization and in vitro digestion. Food Chemistry, 2019, 276: 307–314. https://doi.org/10.1016/j.foodchem.2018.10.026

[47]

Elzoghby, A. O., Samy, W. M., Elgindy, N. A., et al. Protein-based nanocarriers as promising drug and gene delivery systems. Journal of Controlled Release, 2012, 161: 38–49. https://doi.org/10.1016/j.jconrel.2012.04.036

[48]

Ma, L., Su, C. R., Li, S. Y., et al. Co-delivery of curcumin and quercetin in the bilayer structure based on complex coacervation. Food Hydrocolloids, 2023, 144: 108907. https://doi.org/10.1016/j.foodhyd.2023.108907

[49]

Cheng, J. J., Lu, K. Y., Zou, Y., et al. Bi-functional quercetin/copper nanoparticles integrating bactericidal and anti-quorum sensing properties for preventing the formation of biofilms. Biomaterials Science, 2024, 12: 1788. https://doi.org/10.1039/d4bm00034j

[50]

Guan, J. H., Tan, X., Jiao, J., et al. Iron ion-coordinated carrier-free supramolecular co-nanoassemblies of dual DNA topoisomerase-targeting inhibitors for tumor suppression. Acta Biomaterialia, 2022, 144: 121–131. https://doi.org/10.1016/j.actbio.2022.03.027

[51]

Huo, M. R., Zhou, J. Y., Wang, H. L., et al. A pH sensitive nanodrug for collaborative penetration and inhibition of metastatic tumors. Journal of Controlled Release, 2022, 352: 893–908. https://doi.org/10.1016/j.jconrel.2022.11.012

[52]

Cheng, J. J., Wang, S., Han, Y., et al. Carrier-free triterpene prodrugs with glutathione response and biosafety for synergistically enhanced photochemotherapy. ACS Applied Materials & Interfaces, 2021, 13: 245–256. https://doi.org/10.1021/acsami.0c19214

[53]

Cheng, J. J., Wang, J. C., Han, Y., et al. Bioactive natural small molecule-tuned coassembly of photosensitive drugs for highly efficient synergistic and enhanced type I photochemotherapy. ACS Applied Materials & Interfaces, 2020, 12: 43488–43500. https://doi.org/10.1021/acsami.0c13164

[54]

Wang, J. C., Qiao, W. S., Cheng, J. J., et al. Nanomedicine-Cum-Carrier by co-assembly of natural small products for synergistic enhanced antitumor with tissues protective actions. ACS Applied Materials & Interfaces, 2020, 12: 42537–42550. https://doi.org/10.1021/acsami.0c12641

[55]

Li, Z. Y., Shi, H. F., Xie, H. Z., et al. Convenient tuning of the elasticity of self-assembled nano-sized triterpenoids to regulate their biological activities. ACS Applied Materials & Interfaces, 2021, 13: 44065–44078. https://doi.org/10.1021/acsami.1c12418

[56]

Chen, M. Y., Chen, N., Yang, E. L., et al. Co-assemblies based on natural Hemslecin A and β-sitosterol as a new sight for synergistic anti-gastric cancer efficacy in TCM. Colloid and Interface Science Communications, 2022, 49: 100629. https://doi.org/10.1016/j.colcom.2022.100629

[57]

Wei, J., Wang, G., Chen, F., et al. Sol–Gel synthesis of metal–phenolic coordinationspheres and their derived carbon composites. Angewandte Chemie International Edition, 2018, 57: 9838–9843. https://doi.org/10.1002/anie.201805781

[58]

Wang, Z., Li, Y. W., Cheng, Y. Y., et al. Metal-Containing polydopamine nanomaterials: catalysis, energy, and theranostics. Small, 2020, 16: 1907042. https://doi.org/10.1002/smll.201907042

[59]

Zhang, S. C., Lv, X. Y., Dai, H. M., et al. A H2O2 self-sufficient nanoplatform with domino effects for thermal-responsive enhanced chemodynamic therapy. Chemical Science, 2020, 11: 1926. https://doi.org/10.1039/C9SC05506A

[60]

Ren, Z. G., Sun, R. R., Cui, G. Y., et al. A metal polyphenol-coordinated nanomedicine for synergistic cascade cancer chemotherapy. Advanced Materials, 2020, 32: 1906024. https://doi.org/10.1002/adma.201906024

[61]

Liu, T., Liu, W. L., Zeng, X., et al. Metal ion/tannic acid assembly as a versatile photothermal platform in engineering multimodal nanotheranostics for advanced applications. ACS Nano, 2018, 12: 3917–3927. https://doi.org/10.1021/acsnano.8b01456

[62]

Wang, X. Y., Zhang, C. L., Chen, J., et al. High efficient anti-cancer drug delivery systems using tea polyphenols reduced and functionalized graphene oxide. Journal of Biomaterials Applications, 2017, 31: 1108–1122. https://doi.org/10.1177/0885328216689364

[63]

Zhang, M. J., Chen, Y. D., Li, L., et al. Precise synthesis of unique polydopamine/mesoporous calcium phosphate hollow Janus nanoparticles for imaging-guided chemophotothermal synergistic therapy. Chemical Science, 2017, 8: 8067. https://doi.org/10.1039/C7SC03521G

[64]

Zhang, Y., Wang, L., Liu, L., et al. Engineering metal−organic frameworks for photoacoustic imaging-guided chemo-photothermal combinational tumor therapy. ACS Applied Materials & Interfaces, 2018, 10: 41035–41045. https://doi.org/10.1021/acsami.8b13492

[65]

Guo, Y. X., Wu, F. G., Dai, Y. L., et al. Polyphenol-Containing nanoparticles: synthesis, properties, and therapeutic delivery. Advanced Materials, 2021, 33: 2007356. https://doi.org/10.1002/adma.202007356

[66]

Zhang, P. F., Chen, H., Zhao, L., et al. Tumor microenvironment-responsive ultrasmall nanodrug generators with enhanced tumor delivery and penetration. Journal of the American Chemical Society, 2018, 140: 14980–14989. https://doi.org/10.1021/jacs.8b09396

[67]

Pimentel-Moral, S., Teixeira, M. C., Fernandes, A. R., et al. Lipid nanocarriers for the loading of polyphenols–a comprehensive review. Advances in Colloid and Interface Science, 2018, 260: 85–94. https://doi.org/10.1016/j.cis.2018.08.007

[68]

Li, Y., Yang, L. N., Zhao, Y. T., et al. Recent advances in the development and antimicrobial applications of metal–phenolic networks. Advanced Science, 2022, 9: 2202684. https://doi.org/10.1002/advs.202202684

[69]

Jia, Z. J., Cheng, Y., Zheng, Y. F., et al. Strategic advances in spatiotemporal control of bioinspired phenolic chemistries in materials science. Advanced Functional Materials, 2021, 31: 2008821. https://doi.org/10.1002/adfm.202008821

[70]

Shpigelman, A., Israeli, G., Livney, Y. D., et al. Thermally-induced proteinepolyphenol co–assemblies: beta lactoglobulin-based nanocomplexes as protective nanovehicles for EGCG. Food Hydrocolloids, 2010, 24: 735–743. https://doi.org/10.1016/j.foodhyd.2010.03.015

[71]

Han, Y. Y., Zhou, J. J., Xu, W. J., et al. Role of molecular interactions in supramolecular polypeptide−polyphenol networks for engineering functional materials. Journal of the American Chemical Society, 2022, 144: 12510–12519. https://doi.org/10.1021/jacs.2c05052

[72]

Feng, Y. Y., Wei, J. Engineering functional mesoporous materials from plant polyphenol based coordination polymers. Coordination Chemistry Reviews, 2022, 468: 214649. https://doi.org/10.1016/j.ccr.2022.214649

[73]
Dowdy, S. F. Overcoming cellular barriers for RNA therapeutics. Nature Biotechnology, 2017 , 35: 222–229. https://doi.org/10.1038/nbt.3802
[74]

Han, J. P., Cui Y. C., Li, F., et al. Responsive disassembly of nucleic acid nanocomplex in cells for precision medicine. Nano Today, 2021, 39: 101160. https://doi.org/10.1016/j.nantod.2021.101160

[75]

Qin, J. J., Yang, M., Wang, Y. C., et al. Interaction between caffeic acid/caffeic acid phenethyl ester and micellar casein. Food Chemistry, 2021, 349: 129154. https://doi.org/10.1016/j.foodchem.2021.129154

[76]

Yuan, Y. T., Tan, W. Q., Lin, C. H., et al. Development of antioxidant chitosan-based films incorporated with chitooligosaccharide-caffeic acid conjugates. Food Hydrocolloids, 2023, 138: 108431. https://doi.org/10.1016/j.foodhyd.2022.108431

[77]

Jia, F., Li, J., Cai, F. H., et al. Supramolecular nano-assembly of caffeate-strengthened phenylboronic ester with multistep ROS scavenging ability for targeted therapy of acute kidney injury. Advanced Healthcare Materials, 2023, 12: 2301615. https://doi.org/10.1002/adhm.202301615

[78]

Zheng, Y. H., You, X. R., Guan, S. Y., et al. Poly (ferulic acid) with an anticancer effect as a drug nanocarrier for enhanced colon cancer therapy. Advanced Functional Materials, 2019, 29: 1808646. https://doi.org/10.1002/adfm.201808646

[79]

Xia, Y., Lu, G. Z., Li, C. L., et al. Natural phytochemicals in bladder cancer prevention and therapy. Frontiers in Oncology, 2021, 11: 652033. https://doi.org/10.3389/fonc.2021.652033

[80]

Liu, S. S., Li, Y., Wu, C. J., et al. Paeoniflorin suppresses the apoptosis and inflammation of human coronary artery endothelial cells induced by oxidized low-density lipoprotein by regulating the Wnt/ β-catenin pathway. Pharmaceutical Biology, 2023, 61: 1454–1461. https://doi.org/10.1080/13880209.2023.2220360

[81]

Li, G. T., Zheng, S. Z., Xu, L., et al. Zwitterion-driven shape program of prodrug nanoassemblies with high stability, high tumor accumulation, and high antitumor activity. Advanced Healthcare Materials, 2021, 10: 2101407. https://doi.org/10.1002/adhm.202101407

[82]

Huang, X. M., Wang, P. L., Li, T., et al. Self-Assemblies based on traditional medicine berberine and cinnamic acid for adhesion-induced inhibition multidrug-resistant staphylococcus aureus. Applied Materials & Interfaces, 2020, 12: 227–237. https://doi.org/10.1021/acsami.9b17722

[83]

Zheng, T. T., Cui, M. Y., Chen, H., et al. Co-assembled nanocomplexes comprising epigallocatechin gallate and berberine for enhanced antibacterial activity against multidrug resistant staphylococcus aureus. Biomedicine & Pharmacotherapy, 2023, 163: 114856. https://doi.org/10.1016/j.biopha.2023.114856

[84]

Zhou, Z. J., Zhang,Q. Y., Yu, G. C., et al. Exquisite vesicular nanomedicine by paclitaxel mediated co-assembly with camptothecin prodrug. Angewandte Chemie International Edition, 60, 2021, 60: 21033–21039. https://doi.org/10.1002/anie.202108658

[85]

Wang, Z. J., Wang, Y. H., Wang, C. L., et al. Dynamic adjust of non-radiative and radiative attenuation of AIE molecules reinforces NIR-II imaging mediated photothermal therapy and immunotherapy. Advanced Science, 2022, 9: 2104793. https://doi.org/10.1002/advs.202104793

[86]
Desiraju, G. R. A bond by any other name. Angewandte Chemie International Edition, 2011 , 50: 52-59. https://doi.org/10.1002/anie.201002960
[87]

Johnson, E. R., Keinan, S., Mori-Sa´nchez, P., et al. Revealing noncovalent interactions. Journal of the American Chemical Society, 2010, 132: 6498–6506. https://doi.org/10.1021/ja100936w

[88]

Leonard J. P., David N. R., Timmerman, P., et al. Noncovalent synthesis using hydrogen bonding. Angewandte Chemie International Edition, 2001, 40: 2382–2426 https://doi.org/10.1002/1521-3773(20010702)40:13

[89]

Liu, S., Xu, X. P., Ye, J., et al. Metal-coordinated nanodrugs based on natural products for cancer theranostics. Chemical Engineering Journal, 2023, 453: 140892. https://doi.org/10.1016/j.cej.2022.140892

[90]

Han, J. J., Zhao, L. Y., Kim, G., et al. Albumin-mediated “Unlocking” of supramolecular prodrug-like nanozymes toward selective imagingguided phototherapy. Chemical Science, 2022, 13: 7814. https://doi.org/10.1039/D2SC02025D

[91]

Li, K., Richardson, J. J., Tardy, B. L., et al. Targeted therapy against metastatic melanoma based on self-assembled metal-phenolic nanocomplexes comprised of green tea catechin. Advanced Science, 2019, 6: 1801688. https://doi.org/10.1002/advs.201801688

[92]

Wang, Y. A., Yang, Q., Jiang, Y. X., et al. Ovalbumin amyloid-like fibrils/resveratrol self-assembling hydrogel: preparation, characterization and formation mechanism. Food Hydrocolloids, 2024, 151: 109798. https://doi.org/10.1016/j.foodhyd.2024.109798

[93]

Chen, J. Q., Zhou, J. J., Lin, Z. X., et al. Assembly of bioactive nanoparticles via metal–phenolic complexation. Advanced Materials, 2022, 34: 2108624. https://doi.org/10.1002/adma.202108624

[94]

Suna, M., Liu, T., Tan, X., et al. Structurally defined tandem-responsive nanoassemblies composed of dipeptide-based photosensitive derivatives and hypoxia-activated camptothecin prodrugs against primary and metastatic breast tumors. Acta Pharmaceutica Sinica B, 2022, 12: 952–966. https://doi.org/10.1016/j.apsb.2021.08.008

[95]

Huang, H. B., Hou, Y. Y., He, W. Y., et al. Self-assembly of chlorogenic acid into hydrogel for accelerating wound healing. Colloids and Surfaces B: Biointerfaces, 2023, 228: 113440. https://doi.org/10.1016/j.colsurfb.2023.113440

[96]

Li, Y., Xu, Z. H., Tang, L. Q., et al. Nanofibers fortified with synergistic defense route: a potent wound dressing against drug-resistant bacterial infections. Chemical Engineering Journal, 2023, 475: 146492. https://doi.org/10.1016/j.cej.2023.146492

Food & Medicine Homology
Cite this article:
Miao Y-H, Wang X, Zhao X-M, et al. Co-assembly strategies of natural plant compounds for improving their bioavailability. Food & Medicine Homology, 2025, https://doi.org/10.26599/FMH.2025.9420022

949

Views

341

Downloads

0

Crossref

Altmetrics

Received: 04 May 2024
Revised: 10 June 2024
Accepted: 13 June 2024
Published: 18 July 2024
© National R & D Center for Edible Fungus Processing Technology 2025. Published by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return