1
|
Huang R, Wu DX, Wu R, Yang N, Wei F, Dai SY, Zheng J. Research on chemical constituent differences in wild and cultivated Arnebia euchroma by combining UPLC-QTOF-MS with chemometrics. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:3579-3589. [PMID: 40259831 DOI: 10.1039/d4ay01998a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
In this study, ultra-high performance liquid chromatography with quadrupole-time-of-flight mass spectrometry (UPLC-QTOF-MS) was used to analyze the wild and cultivated Arnebia euchroma and the chemical constituent differences were determined by chemometric analysis including principal component analysis (PCA), hierarchical cluster analysis (HCA), and orthogonal partial least squares-discrimination analysis (OPLS-DA). The MS data were processed using UNIFI combined with an in-house library to characterize the metabolites automatically. Based on the multiple adduct ions, exact mass, diagnostic fragment ions, peak intensity, retention time of compounds, and the fragmentation mechanism and retention behavior of references, the structures identified by UNIFI were further verified and the unidentified compounds were also tentatively elucidated. 68 compounds were identified, including 26 shikonins, 14 shikonofurans, 7 flavonoids, 7 phenolic acids, 5 esters, 2 phenylpropanoids, 2 saccharides and 5 others. The chemometrics results showed that the compositions of wild and cultivated Arnebia euchroma were significantly different; 8 differential components were obtained through VIP >1 screening. The quality of wild and cultivated products is strongly influenced by altitude, and the higher the altitude, the more favorable the accumulation of chemical constituents, especially shikonofurans. These results provide basic information for revealing the effects of the ecological environment on the metabolite synthesis and accumulation of Arnebia euchroma and provide an important basis for further research and clinical application of wild and cultivated Arnebia euchroma.
Collapse
Affiliation(s)
- R Huang
- National Institutes for Food and Drug Control, Beijing 102629, China.
| | - D X Wu
- National Institutes for Food and Drug Control, Beijing 102629, China.
| | - R Wu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - N Yang
- The Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - F Wei
- National Institutes for Food and Drug Control, Beijing 102629, China.
| | - S Y Dai
- National Institutes for Food and Drug Control, Beijing 102629, China.
| | - J Zheng
- National Institutes for Food and Drug Control, Beijing 102629, China.
| |
Collapse
|
2
|
Biswal S, Sahoo SK, Biswal BK. Shikonin a potent phytotherapeutic: a comprehensive review on metabolic reprogramming to overcome drug resistance in cancer. Mol Biol Rep 2025; 52:347. [PMID: 40156720 DOI: 10.1007/s11033-025-10459-6] [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: 11/28/2024] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Drug resistance remains a major challenge in cancer therapy, often leading to treatment failure. Metabolic reprogramming, a hallmark of cancer, plays a pivotal role in drug resistance. Phytocompounds, particularly shikonin, a naphthoquinone derived from Lithospermum erythrorhizon, have garnered significant interest as potential alternatives for cancer prevention and treatment. This review focuses on the anticancer properties of shikonin, particularly its ability to modulate metabolic reprogramming and overcome drug resistance. This review, based on extensive searches in databases like PubMed, Web of Science, Google Scholar, and Scopus, highlights shikonin's potential as a therapeutic agent. Shikonin exhibits a wide range of anticancer activities, including induction of apoptosis, autophagy, necroptosis, inhibition of angiogenesis, invasion, and migration, as well as disruption of the cell cycle and promotion of DNA damage. It targets altered cancer cell metabolism to inhibit proliferation and reverse drug resistance, making it a promising candidate for therapeutic development. Preliminary clinical trials suggest that shikonin can enhance the efficacy of established chemotherapeutic agents, immunotherapies, and radiation through additive and synergistic interactions. Despite its promise, further research is needed to elucidate the precise mechanisms underlying shikonin's metabolic reprogramming effects in cancer. A comprehensive understanding could pave the way for its integration into standard oncological treatments. With its capacity to act on multiple cancer pathways and enhance conventional treatments, shikonin stands out as a viable candidate for combating drug-resistant cancers and advancing clinical oncology.
Collapse
Affiliation(s)
- Stuti Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | | | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
3
|
Cao S, Li H, Ye X, Xing X, Xie Y, Zeng X, Liu H, Zhong X, Yang X, Xing W, Zhu C, Wu X. Shikonin induces the apoptosis and pyroptosis of EGFR-T790M-mutant drug-resistant non-small cell lung cancer cells via the degradation of cyclooxygenase-2. Eur J Med Res 2024; 29:611. [PMID: 39702296 DOI: 10.1186/s40001-024-02187-7] [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: 09/13/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND The T790M mutation in the epidermal growth factor receptor (EGFR) gene is the primary cause of resistance to EGFR-tyrosine kinase inhibitor (TKI) therapy in non-small cell lung cancer (NSCLC) patients. Previous research demonstrated that certain traditional Chinese medicine (TCM) monomers exhibit anti-tumor effects against various malignancies. This study aims to investigate the potentials of shikonin screened from a TCM monomer library containing 1060 monomers in killing EGFR-T790M drug-resistant NSCLC cells and elucidate the underlying mechanisms. METHODS MTT method was used to screen for the TCM monomers with significant killing effects on H1975 cells carrying the EGFR-T790M mutation. The influences of the identified monomer shikonin on cell growth were determined by the colony formation assay. Annexin-V/PI staining and JC-1 staining were applied to detect the effects of shikonin on cell apoptosis. The influences of shikonin on cell membrane integrity were detected by lactate dehydrogenase (LDH) release assay. Reactive oxygen species (ROS) generation was analyzed using DCFH-DA as probe. The mechanisms of shikonin affecting the stability of cyclooxygenase-2 (COX-2) were evaluated by using specific inhibitors for protein degradation pathways. Western blotting was performed to assess the effects of the alteration of COX-2 expression or enzymatic activity on the related signal pathways as well as the apoptotic and pyroptotic markers. RESULTS Shikonin was identified as a potent cytotoxic compound against EGFR-T790M-mutant NSCLC cells. Shikonin induced cell apoptosis and pyroptosis by triggering the activation of the caspase cascade and cleavage of poly (ADP-ribose) polymerase and gasdermin E by elevating intracellular ROS levels. Further investigations revealed that shikonin induced the degradation of COX-2 via the proteasome pathway, thereby decreasing COX-2 protein level and enzymatic activity and subsequently inhibiting the downstream PDK1/Akt and Erk1/2 signaling pathways through the induction of ROS production. Notably, COX-2 overexpression attenuated shikonin-induced apoptosis and pyroptosis, whereas COX-2 inhibition with celecoxib enhanced the cytotoxic effects of shikonin. CONCLUSIONS Combination treatment with shikonin and COX-2 inhibitor may be a suitable therapeutic strategy for EGFR-T790M-mutant NSCLC treatment.
Collapse
Affiliation(s)
- Shaoyi Cao
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Huaqiu Li
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Xiaoyan Ye
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Xinxing Xing
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Yonghuan Xie
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Xiangfeng Zeng
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Hongjiao Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Zhujiang New Town, Tianhe, Guangzhou, 510623, China
| | - Xing Zhong
- The First Clinical Medical College, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xinyi Yang
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Wenxiu Xing
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China
| | - Cairong Zhu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Zhujiang New Town, Tianhe, Guangzhou, 510623, China.
| | - Xiaoping Wu
- Department of Immunology and Microbiology, College of Life Science and Technology, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, No. 601 Huangpu Avenue West, Tianhe, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Cen LS, Cao Y, Zhou YM, Guo J, Xue JW. Shikonin protects mitochondria through the NFAT5/AMPK pathway for the treatment of diabetic wounds. World J Diabetes 2024; 15:2338-2352. [PMID: 39676806 PMCID: PMC11580590 DOI: 10.4239/wjd.v15.i12.2338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Shikonin is a natural remedy that is effective at treating diabetic wounds. NFAT5 is a potential therapeutic target for diabetes, and mitochondrial function is essential for wound healing. However, the relationship among Shikonin, NFAT5, and mitochondrial function has not been thoroughly studied. Here, we offer new perspectives on the advantages of shikonin for managing diabetes. AIM To assess the therapeutic mechanism of shikonin in diabetic wounds, its relationship with NFAT5, and its protection of mitochondrial function. METHODS Hypertonic cell and diabetic wound mouse models were established. NFAT5 expression was measured through western blotting and immunofluorescence, in vivo and in vitro. Mitochondrial function was evaluated using reactive oxygen species (ROS) detection and JC-1 and Calcein AM dyes. Mitochondrial structures were observed using transmission electron microscopy. The NFAT5/AMPK pathway was analyzed using a transfection vector and an inhibitor. The effect of shikonin on cells under hypertonic conditions via the NFAT5/AMPK pathway was assessed using western blotting. RESULTS Shikonin treatment preserved HaCaT cell viability, while significantly reducing cyclooxygenase-2 expression levels in a high-glucose environment (P < 0.05). Additionally, shikonin maintained mitochondrial morphology, enhanced membrane potential, reduced membrane permeability, and decreased ROS levels in HaCaT cells under hyperosmolar stress. Furthermore, shikonin promoted wound healing in diabetic mice (P < 0.05). Shikonin also inhibited NFAT5, in vivo and in vitro (P < 0.05). Shikonin treatment reduced NFAT5 expression levels, subsequently inhibiting AMPK expression in vitro (P < 0.05). Finally, shikonin inhibited several key downstream molecules of the NFAT5/AMPK pathway, including mammalian target of rapamycin, protein kinase B, nuclear factor kappa-light-chain-enhancer of activated B cells, and inducible nitric oxide synthase (P < 0.05). CONCLUSION Shikonin protects mitochondria via the NFAT5/AMPK-related pathway and enhances wound healing in diabetes.
Collapse
Affiliation(s)
- Lu-Sha Cen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Yi-Mai Zhou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Jing Guo
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Jing-Wen Xue
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, Zhejiang Province, China
| |
Collapse
|
5
|
Lu C, Zhang Z, Fan Y, Wang X, Qian J, Bian Z. Shikonin induces ferroptosis in osteosarcomas through the mitochondrial ROS-regulated HIF-1α/HO-1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156139. [PMID: 39423479 DOI: 10.1016/j.phymed.2024.156139] [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: 05/29/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The most common malignant bone tumour is osteosarcoma, which has an unsatisfactory prognosis and unsatisfactory treatment. Ferroptosis shows promise as an effective OS therapy. A substance extracted from the Lithospermum erythrorhizon, Shikonin (SHK), inhibits a number of tumours, including ovarian, gastric, and lung cancers. However, whether SHK induces OS ferroptosis and its mechanisms are not clear. PURPOSE Our study is aimed at investigating whether SHK causes ferroptosis and elucidating its molecular mechanism. METHODS Cell counting Kit-8, cell cycle and cell apoptosis assay were utilised to assess therapeutic effect of SHK against OS. Normal cells, including H9C2, AML-12 and HK-2, haematoxylin and eosin staining and mice serum biomarker tests were used to assess SHK biosafety. Malondialdehyde (MDA) levels, the glutathione (GSH)/oxidized glutathione (GSSG) ratio, reactive oxygen species (ROS), lipid peroxide (LPO), and intracellular Fe2+ detection, quantitative reverse transcription PCR (qRT-PCR), Western blotting (WB) and rescue experiments were employed to confirm whether SHK induced ferroptosis in OS cells. Molecular docking, cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS) assay were used to evaluate the direct binding between SHK and hypoxia-inducible factor-1α (HIF-1α). Protein stability and degradation analysis, small RNA interference, flow cytometry, qRT-PCR, and WB were used to investigate the molecular mechanism of ferroptosis. In vivo xenografts of nude mouse was used to study the anti-OS effect of SHK. RESULTS SHK significantly reduced OS cell viability and induced apoptosis and G2/M arrest. SHK increased intracellular levels of MDA, ROS, LPO, and Fe2+ while simultaneously reducing the GSH/GSSG ratio and GPX4 and SLC7A11 expression. CETSA and DARTS results demonstrated that SHK did not bind targetly to HIF-1α. Instead, mitochondrial ROS (MitoROS) promoted HIF-1α expression, resulting in HO-1 overexpression, excess Fe2+ production, ROS accumulation and GPX depletion, and ferroptosis. Furthermore, inhibition of MitoROS using Mito-TEMPO downregulated HIF-1α/HO-1 axis and mitigated the SHK-induced ferroptosis. In vivo, SHK effectively suppressed OS growth with favourable biosafety, confirming the molecular mechanism underlying SHK-induced ferroptosis in OS. CONCLUSION We observe that HIF-1α/HO-1 axis is the crucial factor in SHK-induced OS ferroptosis. Importantly, we demonstrate that HIF-1α is indirectly regulated by MitoROS rather than SHK bound directly to HIF-1α. Our research suggest that SHK is a potential drug candidate for OS treatment and may help in identifying novel therapeutic targets for OS.
Collapse
Affiliation(s)
- Congcong Lu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China; Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Zhen Zhang
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Yuhao Fan
- Zhejiang University School of Medicine, Hangzhou, 310000, PR China
| | - Xiyu Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China; Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Jin Qian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China.
| | - Zhenyu Bian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China.
| |
Collapse
|
6
|
Xue S, Lin Y, Chen H, Yang Z, Zha J, Jiang X, Han Z, Wang K. Mechanisms of autophagy and their implications in dermatological disorders. Front Immunol 2024; 15:1486627. [PMID: 39559368 PMCID: PMC11570406 DOI: 10.3389/fimmu.2024.1486627] [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: 08/26/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
Autophagy is a highly conserved cellular self-digestive process that underlies the maintenance of cellular homeostasis. Autophagy is classified into three types: macrophage, chaperone-mediated autophagy (CMA) and microphagy, which maintain cellular homeostasis through different mechanisms. Altered autophagy regulation affects the progression of various skin diseases, including psoriasis (PA), systemic lupus erythematosus (SLE), vitiligo, atopic dermatitis (AD), alopecia areata (AA) and systemic sclerosis (SSc). In this review, we review the existing literature focusing on three mechanisms of autophagy, namely macrophage, chaperone-mediated autophagy and microphagy, as well as the roles of autophagy in the above six dermatological disorders in order to aid in further studies in the future.
Collapse
Affiliation(s)
- Shenghao Xue
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Haoran Chen
- Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhengyu Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Junting Zha
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xuan Jiang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Zhongyu Han
- Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Ke Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| |
Collapse
|
7
|
Zhang Y, Chen L, Ouyang H. Shikonin alleviates asthma phenotypes in mice via an airway epithelial STAT3-dependent mechanism. Open Med (Wars) 2024; 19:20241016. [PMID: 39444792 PMCID: PMC11497215 DOI: 10.1515/med-2024-1016] [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: 02/19/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 10/25/2024] Open
Abstract
Background Asthma is an inflammatory disease where the balance between Th1/Th2 and Th17/Treg plays a crucial role in its pathogenesis. Shikonin is used to treat a variety of autoimmune diseases due to its good anti-inflammatory activity. However, the effect and mechanism of shikonin on asthma remain unknown. Method Mice were sensitized with ovalbumin (OVA)/house dust mite (HDM) and treated with shikonin. Lung inflammation was assessed histologically and via flow cytometry. Bronchoalveolar lavage fluid (BALF) was analyzed for cell counts and cytokines. Shikonin's impact on p-STAT3 was studied in vivo and in vitro. Results Shikonin inhibited OVA or HDM-induced inflammation and airway hyperresponsiveness. Upon treatment, a restoration of the Th1/Th2 and Th17/Treg balance was observed, evidenced by a reduction in IL-4 and IL-17A levels in BALF, alongside an elevation in interferon-gamma and IL-10. Furthermore, shikonin impeded the infiltration of eosinophils, neutrophils, macrophages, and lymphocytes into lung tissue. The observed decrease in STAT3 phosphorylation and diminished nuclear translocation of p-STAT3 confirmed that shikonin promotes the balance of Th1/Th2 and Th17/Treg by regulating airway epithelial STAT3. Conclusion Shikonin mitigates asthma symptoms through a STAT3-dependent mechanism, indicating its potential as an anti-asthmatic therapeutic agent.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Respiratory Medicine, Xi’an International Medical Center Hospital, Xi’an, 710101, China
| | - Lizhan Chen
- Department of Respiratory Medicine, Xi’an International Medical Center Hospital, Xi’an, 710101, China
| | - Haifeng Ouyang
- Department of Respiratory Medicine, Xi’an International Medical Center Hospital, No. 777 Xitai Road, Xi’an, 710101, China
| |
Collapse
|
8
|
Chen L, Xu YX, Wang YS, Ren YY, Chen YM, Zheng C, Xie T, Jia YJ, Zhou JL. Integrative Chinese-Western medicine strategy to overcome docetaxel resistance in prostate cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118265. [PMID: 38677579 DOI: 10.1016/j.jep.2024.118265] [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: 03/15/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicines (TCMs) have emerged as a promising complementary therapy in the management of prostate cancer (PCa), particularly in addressing resistance to Docetaxel (DTX) chemotherapy. AIM OF THE REVIEW This review aims to elucidate the mechanisms underlying the development of resistance to DTX in PCa and explore the innovative approach of integrating TCMs in PCa treatment to overcome this resistance. Key areas of investigation include alterations in microtubule proteins, androgen receptor and androgen receptor splice variant 7, ERG rearrangement, drug efflux mechanisms, cancer stem cells, centrosome clustering, upregulation of the PI3K/AKT signaling pathway, enhanced DNA damage repair capability, and the involvement of neurotrophin receptor 1/protein kinase C. MATERIALS AND METHODS With "Prostate cancer", "Docetaxel", "Docetaxel resistance", "Natural compounds", "Traditional Chinese medicine", "Traditional Chinese medicine compound", "Medicinal plants" as the main keywords, PubMed, Web of Science and other online search engines were used for literature retrieval. RESULTS Our findings underscore the intricate interplay of molecular alterations that collectively contribute to the resistance of PCa cells to DTX. Moreover, we highlight the potential of TCMs as a promising complementary therapy, showcasing their ability to counteract DTX resistance and enhance therapeutic efficacy. CONCLUSION The integration of TCMs in PCa treatment emerges as an innovative approach with significant potential to overcome DTX resistance. This review not only provides insights into the mechanisms of resistance but also presents new prospects for improving the clinical outcomes of patients with PCa undergoing DTX therapy. The comprehensive understanding of these mechanisms lays the foundation for future research and the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yi-Min Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Cheng Zheng
- Department of Traditional Chinese Medicines, Zhejiang Institute for Food and Drug Control, Hangzhou, Zhejiang 310052, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Ying-Jie Jia
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
9
|
Shi C, Qi Z, Yang C, Luo S, Huang S, Luo Y. Shikonin ameliorates depressive- and anxiogenic-like behaviors in rats via the suppression of inflammation in the hippocampus. Neurosci Lett 2024; 837:137893. [PMID: 38997082 DOI: 10.1016/j.neulet.2024.137893] [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: 02/13/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Shikonin is an active naphthoquinone with antioxidative, anti-inflammatory, and anticancer properties. In this study, we investigated the effects of shikonin on depressive- and anxiety-like behaviors in lipopolysaccharide- (LPS-) induced depression and chronic unpredictable mild stress (CUMS) rat models and explored the potential mechanism. First, a 14-day intraperitoneal administration of shikonin (10 mg/kg) significantly decreased immobility time in forced swimming test (FST) and increased open arm entries in elevated plus maze (EPM) test, without affecting line crossings in open field test (OFT), indicating that shikonin has anti-depressant- and anxiolytic-like effects. Second, chronic shikonin administration (10 mg/kg) reversed depressive- and anxiety-like behaviors in LPS-induced and CUMS depression models, as shown in the sucrose preference test (SPT), FST, EPM, and novel object recognition test (NORT). Finally, shikonin significantly reduced the levels of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α) in hippocampus, indicating that the anti-depressant- and anxiolytic-like effects of shikonin are related to the reduction of neuroinflammation in hippocampus. These findings suggest that shikonin exerts anti-depressant- and anxiolytic-like effects via an anti-inflammatory mechanism of shikonin in the hippocampus.
Collapse
Affiliation(s)
- Cuijie Shi
- Department of Pharmacology, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Zihan Qi
- Department of Pharmacology, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Chang Yang
- Department of Pharmacology, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Shuting Luo
- Department of Pharmacology, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191 Beijing, China.
| | - Yixiao Luo
- Department of Pharmacology, School of Medicine, Hunan Normal University, Changsha 410013, China; Hunan Province People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
10
|
Liu T, Zheng N, Ma Y, Zhang Y, Lei H, Zhen X, Wang Y, Gou D, Zhao J. Recent advancements in chitosan-based intelligent food freshness indicators: Categorization, advantages, and applications. Int J Biol Macromol 2024; 275:133554. [PMID: 38950804 DOI: 10.1016/j.ijbiomac.2024.133554] [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: 04/23/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
With an increasing emphasis on food safety and public health, there is an ongoing effort to develop reliable, non-invasive methods to assess the freshness of diverse food products. Chitosan-based food freshness indicators, leveraging properties such as biocompatibility, biodegradability, non-toxicity, and high stability, offer an innovative approach for real-time monitoring of food quality during storage and transportation. This review introduces intelligent food freshness indicators, specifically those utilizing pH-sensitive dyes like anthocyanins, curcumin, alizarin, shikonin, and betacyanin. It highlights the benefits of chitosan-based intelligent food freshness indicators, emphasizing improvements in barrier and mechanical properties, antibacterial activity, and composite film solubility. The application of these indicators in the food industry is then explored, alongside a concise overview of chitosan's limitations. The paper concludes by discussing the challenges and potential areas for future research in the development of intelligent food freshness indicators using chitosan. Thus, chitosan-based smart food preservation indicators represent an innovative approach to providing real-time data for monitoring food quality, offering valuable insights to both customers and retailers, and playing a pivotal role in advancing the food industry.
Collapse
Affiliation(s)
- Tong Liu
- College of Food Science and Engineering, Changchun University, Changchun 130022, China; Key Laboratory of Intelligent Rehabilitation and Barrier-free for the Disabled Ministry of Education, Changchun University, Changchun 130022, China
| | - Nan Zheng
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Yaomei Ma
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Yu Zhang
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Hongyu Lei
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Xinyu Zhen
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Yue Wang
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Dongxia Gou
- College of Food Science and Engineering, Changchun University, Changchun 130022, China; Key Laboratory of Intelligent Rehabilitation and Barrier-free for the Disabled Ministry of Education, Changchun University, Changchun 130022, China
| | - Jun Zhao
- College of Food Science and Engineering, Changchun University, Changchun 130022, China; Key Laboratory of Intelligent Rehabilitation and Barrier-free for the Disabled Ministry of Education, Changchun University, Changchun 130022, China.
| |
Collapse
|
11
|
Yang C, Ma Y, Lu Q, Qu Y, Li Y, Cheng S, Xiao C, Chen J, Wang C, Wang F, Xiang AP, Huang W, Tang X, Zheng H. 2-Bromo-1,4-Naphthalenedione promotes CD8 + T cell expansion and limits Th1/Th17 to mitigate experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:181. [PMID: 39068463 PMCID: PMC11283727 DOI: 10.1186/s12974-024-03172-x] [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: 04/24/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Treating Multiple sclerosis (MS), a well-known immune-mediated disease characterized by axonal demyelination, is challenging due to its complex causes. Naphthalenedione, present in numerous plants, is being explored as a potential medicine for MS due to its immunomodulatory properties. However, its effects on lymphocytes can vary depending on factors such as the specific compound, concentration, and experimental conditions. In this study, we aim to explore the therapeutic potential of 2-bromo-1,4-naphthalenedione (BrQ), a derivative of naphthalenedione, in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, and to elucidate its underlying mechanisms. We observed that mice treated with BrQ exhibited reduced severity of EAE symptoms, including lower clinical scores, decreased leukocyte infiltration, and less extensive demyelination in central nervous system. Furthermore, it was noted that BrQ does not directly affect the remyelination process. Through cell-chat analysis based on bulk RNA-seq data, coupled with validation of flow analysis, we discovered that BrQ significantly promotes the expansion of CD8+ T cells and their interactions with other immune cells in peripheral immune system in EAE mice. Subsequent CD8+ T cell depletion experiments confirmed that BrQ alleviates EAE in a CD8+ T cell-dependent manner. Mechanistically, expanded CD8+ cells were found to selectively reduce antigen-specific CD4+ cells and subsequently inhibit Th1 and Th17 cell development in vivo, ultimately leading to relief from EAE. In summary, our findings highlight the crucial role of BrQ in modulating the pathogenesis of MS, suggesting its potential as a novel drug candidate for treating MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Cuixia Yang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Yuanchen Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiying Lu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Yuliang Qu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yuantao Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
| | - Shimei Cheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Chongjun Xiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Jinshuo Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Chuangjia Wang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Feng Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China.
| | - Xiaorong Tang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, China.
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| |
Collapse
|
12
|
Zhao Q, Wang K, Hou L, Guo L, Liu X. Based on network pharmacology and molecular docking to explore the potential mechanism of shikonin in periodontitis. BMC Oral Health 2024; 24:839. [PMID: 39048977 PMCID: PMC11270799 DOI: 10.1186/s12903-024-04618-7] [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: 04/21/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVES To investigate the potential mechanisms of shikonin in preventing and treating periodontitis using network pharmacology and molecular docking methods. MATERIALS AND METHODS The targets of shikonin were obtained in TCMSP and SEA databases, and targets of periodontitis were gathered from the OMIM, GeneCards and Drugbank Databases. The intersecting targets were entered into the DAVID database to obtain the relevant biological functions and pathways by GO and KEGG enrichment analysis. The obtained targets were analysed the protein-protein interaction (PPI) in STRING platform. In Cytoscape 3.8.0, the network analysis function with the MCODE plug-in were used to obtain the key targets, of shikonin and periodontitis. Molecular docking and molecular dynamics simulation (MD) were used to assess the affinity between the shikonin and the key targets. RESULTS Shikonin was screened for 22 targets and periodontitis was screened for 944 targets, the intersecting targets were considered as potential therapeutic targets. The targets played important roles in cellular response to hypoxia, response to xenobiotic stimulus and positive regulates of apoptotic process by GO enrichment analysis. 10 significant pathways were analyzed by KEGG, such as human cytomegalovirus infection and PI3K-Akt signaling pathway, etc. Cytoscape software screened the key genes including AKT1, CCL5, CXCR4, PPARG, PTEN, PTGS2 and TP53. Molecular docking and MD results showed that shikonin could bind stably to the targets. CONCLUSIONS The present study enriched the molecular mechanisms in periodontitis with shikonin, providing potential therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Qingliang Zhao
- Department of Stomatology, Harbin the First Hospital, Harbin, 150010, China
| | - Kun Wang
- Department of Central Sterile Supply, the First Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Lin Hou
- Department of Stomatology, Harbin the First Hospital, Harbin, 150010, China
| | - Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town,Jinghai District, Tianjin, 301617, China.
| | - Xiangyan Liu
- Department of Stomatology, Harbin the First Hospital, Harbin, 150010, China.
| |
Collapse
|
13
|
Wang Y, Zeng Y, Yang W, Wang X, Jiang J. Targeting CD8 + T cells with natural products for tumor therapy: Revealing insights into the mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155608. [PMID: 38642413 DOI: 10.1016/j.phymed.2024.155608] [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: 02/24/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Despite significant advances in cancer immunotherapy over the past decades, such as T cell-engaging chimeric antigen receptor (CAR)-T cell therapy and immune checkpoint blockade (ICB), therapeutic failure resulting from various factors remains prevalent. Therefore, developing combinational immunotherapeutic strategies is of great significance for improving the clinical outcome of cancer immunotherapy. Natural products are substances that naturally exist in various living organisms with multiple pharmacological or biological activities, and some of them have been found to have anti-tumor potential. Notably, emerging evidences have suggested that several natural compounds may boost the anti-tumor effects through activating immune response of hosts, in which CD8+ T cells play a pivotal role. METHODS The data of this review come from PubMed, Web of Science, Google Scholar, and ClinicalTrials (https://clinicaltrials.gov/) with the keywords "CD8+ T cell", "anti-tumor", "immunity", "signal 1", "signal 2", "signal 3", "natural products", "T cell receptor (TCR)", "co-stimulation", "co-inhibition", "immune checkpoint", "inflammatory cytokine", "hesperidin", "ginsenoside", "quercetin", "curcumin", "apigenin", "dendrobium officinale polysaccharides (DOPS)", "luteolin", "shikonin", "licochalcone A", "erianin", "resveratrol", "procyanidin", "berberine", "usnic acid", "naringenin", "6-gingerol", "ganoderma lucidum polysaccharide (GL-PS)", "neem leaf glycoprotein (NLGP)", "paclitaxel", "source", "pharmacological activities", and "toxicity". These literatures were published between 1993 and 2023. RESULTS Natural products have considerable advantages as anti-tumor drugs based on the various species, wide distribution, low price, and few side effects. This review summarized the effects and mechanisms of some natural products that exhibit anti-tumor effects via targeting CD8+ T cells, mainly focused on the three signals that activate CD8+ T cells: TCR, co-stimulation, and inflammatory cytokines. CONCLUSION Clarifying the role and underlying mechanism of natural products in cancer immunotherapy may provide more options for combinational treatment strategies and benefit cancer therapy, to shed light on identifying potential natural compounds for improving the clinical outcome in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuke Wang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Neurosurgery, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Yan Zeng
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenyong Yang
- Department of Neurosurgery, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xiuxuan Wang
- Research and Development Department, Beijing DCTY Biotech Co., Ltd., Beijing, China
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Fan S, Yan X, Hu X, Liu X, Zhao S, Zhang Y, Zhou X, Shen X, Qi Q, Chen Y. Shikonin blocks CAF-induced TNBC metastasis by suppressing mitochondrial biogenesis through GSK-3β/NEDD4-1 mediated phosphorylation-dependent degradation of PGC-1α. J Exp Clin Cancer Res 2024; 43:180. [PMID: 38937832 PMCID: PMC11210116 DOI: 10.1186/s13046-024-03101-z] [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: 02/29/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by its high metastatic potential, which results in poor patient survival. Cancer-associated fibroblasts (CAFs) are crucial in facilitating TNBC metastasis via induction of mitochondrial biogenesis. However, how to inhibit CAF-conferred mitochondrial biogenesis is still needed to explore. METHODS We investigated metastasis using wound healing and cell invasion assays, 3D-culture, anoikis detection, and NOD/SCID mice. Mitochondrial biogenesis was detected by MitoTracker green FM staining, quantification of mitochondrial DNA levels, and blue-native polyacrylamide gel electrophoresis. The expression, transcription, and phosphorylation of peroxisome-proliferator activated receptor coactivator 1α (PGC-1α) were detected by western blotting, chromatin immunoprecipitation, dual-luciferase reporter assay, quantitative polymerase chain reaction, immunoprecipitation, and liquid chromatography-tandem mass spectrometry. The prognostic role of PGC-1α in TNBC was evaluated using the Kaplan-Meier plotter database and clinical breast cancer tissue samples. RESULTS We demonstrated that PGC-1α indicated lymph node metastasis, tumor thrombus formation, and poor survival in TNBC patients, and it was induced by CAFs, which functioned as an inducer of mitochondrial biogenesis and metastasis in TNBC. Shikonin impeded the CAF-induced PGC-1α expression, nuclear localization, and interaction with estrogen-related receptor alpha (ERRα), thereby inhibiting PGC-1α/ERRα-targeted mitochondrial genes. Mechanistically, the downregulation of PGC-1α was mediated by synthase kinase 3β-induced phosphorylation of PGC-1α at Thr295, which associated with neural precursor cell expressed developmentally downregulated 4e1 recognition and subsequent degradation by ubiquitin proteolysis. Mutation of PGC-1α at Thr295 negated the suppressive effects of shikonin on CAF-stimulated TNBC mitochondrial biogenesis and metastasis in vitro and in vivo. CONCLUSIONS Our findings indicate that PGC-1α is a viable target for blocking TNBC metastasis by disrupting mitochondrial biogenesis, and that shikonin merits potential for treatment of TNBC metastasis as an inhibitor of mitochondrial biogenesis through targeting PGC-1α.
Collapse
Affiliation(s)
- Shuangqin Fan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xiaomin Yan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xiaoxia Hu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xing Liu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Shijie Zhao
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Yue Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xiaofeng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiangchun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China.
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China.
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
| |
Collapse
|
15
|
Lee YG, Jung Y, Choi HK, Lee JI, Lim TG, Lee J. Natural Product-Derived Compounds Targeting Keratinocytes and Molecular Pathways in Psoriasis Therapeutics. Int J Mol Sci 2024; 25:6068. [PMID: 38892253 PMCID: PMC11172960 DOI: 10.3390/ijms25116068] [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: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory skin disorder that affects approximately 2-3% of the global population due to significant genetic predisposition. It is characterized by an uncontrolled growth and differentiation of keratinocytes, leading to the formation of scaly erythematous plaques. Psoriasis extends beyond dermatological manifestations to impact joints and nails and is often associated with systemic disorders. Although traditional treatments provide relief, their use is limited by potential side effects and the chronic nature of the disease. This review aims to discuss the therapeutic potential of keratinocyte-targeting natural products in psoriasis and highlight their efficacy and safety in comparison with conventional treatments. This review comprehensively examines psoriasis pathogenesis within keratinocytes and the various related signaling pathways (such as JAK-STAT and NF-κB) and cytokines. It presents molecular targets such as high-mobility group box-1 (HMGB1), dual-specificity phosphatase-1 (DUSP1), and the aryl hydrocarbon receptor (AhR) for treating psoriasis. It evaluates the ability of natural compounds such as luteolin, piperine, and glycyrrhizin to modulate psoriasis-related pathways. Finally, it offers insights into alternative and sustainable treatment options with fewer side effects.
Collapse
Affiliation(s)
- Yu Geon Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Younjung Jung
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Hyo-Kyoung Choi
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Jae-In Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Tae-Gyu Lim
- Department of Food Science & Biotechnology, College of Life Sciences, Sejong University, Seoul 05006, Republic of Korea;
- Carbohydrate Bioproduct Research Center, Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jangho Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| |
Collapse
|
16
|
Zhang X, Shi Q, Hu M, Zhu K, Zhu L, Cao J, Li C. Holothuria leucospilota polysaccharides (HLP) ameliorate colitis rats via regulation of the metabolic profiling and TLR4/NLRP3 signaling pathways. FOOD FRONTIERS 2024; 5:656-667. [DOI: 10.1002/fft2.343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
AbstractRecently, the development of natural polysaccharides for ameliorating immunity and gut metabolism has attracted extensive attention. This study used Holothuria leucospilota polysaccharides (HLP) to explore the improvement mechanism in ulcerative colitis rats on perspectives of immunity and metabolism. The results showed that HLP increased goblet cells’ number and the content of tight junction proteins (zona occludens 1 and occludin) and improved intestinal barrier permeability. The levels of immune cytokines (IL‐4, IL‐6, IL‐10, IL‐18, TNF‐α, and IL‐1β) and the activity of oxidative stress‐related enzymes (superoxide dismutase, catalase, malondialdehyde, and glutathione peroxidase) were regulated. HLP regulated the related genes and proteins expression of immune cytokines, MAPK, and NLRP3 inflammasome. Furthermore, HLP treatment increased the concentration of short‐chain fatty acids (SCFAs) and regulated serum metabolic disorders by regulating amino acid metabolism, SCFA metabolism, and energy metabolism. These results provide a new perspective for developing HLP as a promising functional food for preventing and mitigating colitis.
Collapse
Affiliation(s)
- Xin Zhang
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering Hainan University Haikou China
| | - Qiuge Shi
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering Hainan University Haikou China
| | - Maojie Hu
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering Hainan University Haikou China
| | - Kexue Zhu
- Spice and Beverage Research Institute Chinese Academy of Tropical Agricultural Sciences Wanning China
| | - Lulu Zhu
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering Hainan University Haikou China
| | - Jun Cao
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering Hainan University Haikou China
| | - Chuan Li
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering Hainan University Haikou China
- Collaborative Innovation Center of Provincial and Ministerial Co‐construction for Marine Food Deep Processing Dalian Polytechnic University Dalian China
| |
Collapse
|